US11173202B2 - Agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (DC-ASGPR) - Google Patents

Agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (DC-ASGPR) Download PDF

Info

Publication number
US11173202B2
US11173202B2 US16/372,832 US201916372832A US11173202B2 US 11173202 B2 US11173202 B2 US 11173202B2 US 201916372832 A US201916372832 A US 201916372832A US 11173202 B2 US11173202 B2 US 11173202B2
Authority
US
United States
Prior art keywords
asgpr
antigen
galβ1
cells
fucα1
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active, expires
Application number
US16/372,832
Other versions
US20190231865A1 (en
Inventor
Sangkon Oh
Dapeng Li
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Baylor Research Institute
Original Assignee
Baylor Research Institute
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Baylor Research Institute filed Critical Baylor Research Institute
Priority to US16/372,832 priority Critical patent/US11173202B2/en
Assigned to BAYLOR RESEARCH INSTITUTE reassignment BAYLOR RESEARCH INSTITUTE ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LI, DAPENG, OH, SANGKON
Publication of US20190231865A1 publication Critical patent/US20190231865A1/en
Priority to US17/454,108 priority patent/US20220152192A1/en
Application granted granted Critical
Publication of US11173202B2 publication Critical patent/US11173202B2/en
Active legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/145Orthomyxoviridae, e.g. influenza virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1027Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against receptors, cell-surface antigens or cell-surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6849Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/37Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from fungi
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/405Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from algae
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0639Dendritic cells, e.g. Langherhans cells in the epidermis
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5154Antigen presenting cells [APCs], e.g. dendritic cells or macrophages
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/74Inducing cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/75Agonist effect on antigen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16111Influenzavirus A, i.e. influenza A virus
    • C12N2760/16122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16111Influenzavirus A, i.e. influenza A virus
    • C12N2760/16134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Definitions

  • the present invention relates in general to the field of agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (DC-ASGPR).
  • DC-ASGPR dendritic cell asialoglycoprotein receptor
  • Dendritic Cells play a pivotal role in controlling the interface of innate and acquired immunity by providing soluble and intercellular signals, followed by recognition of pathogens. These functions of DCs are largely dependent on the expression of specialized surface receptors, ‘pattern recognition receptors’ (PRRs), represented, most notably, by toll-like receptors (TLRs) and C-type lectins or lectin-like receptors (LLRs) (1-3).
  • PRRs pattern recognition receptors
  • TLRs toll-like receptors
  • LLRs lectin-like receptors
  • TLRs interleukin 12
  • IL-12 interleukin 12
  • C-type LLRs operate as constituents of the powerful antigen capture and uptake mechanism of macrophages and DCs (1).
  • LLRs might have broader ranges of biological functions that include cell migrations (4), intercellular interactions (5).
  • These multiple functions of LLRs might be due to the facts that LLRs, unlike TLRs, can recognize both self and non-self.
  • the complexity of LLRs including the redundancy of a number of LLRs expressed in immune cells, has been one of the major obstacles to understand the detailed functions of individual LLRs.
  • natural ligands for most of these receptors remain unidentified. Nonetheless, evidence from recent studies suggests that LLRs, in collaboration with TLRs, may contribute to the activation of immune cells during microbial infections (6-14).
  • DC-ASGPR mRNA was observed predominantly in immune tissues—in DC and granulocytes, but not in T, B, or NK cells, or monocytes.
  • DC-ASGPR species were restricted to the CD14-derived DC obtained from CD34-derived progenitors, while absent from the CD1a-derived subset. Both monocyte-derived DC and tonsillar interstitial-type DC expressed DC-ASGPR protein, while Langerhans-type cells did not.
  • DC-ASGPR was a feature of immaturity, as expression was lost upon CD40 activation.
  • mAb against DC-ASGPR was rapidly internalized by DC at 37° C.
  • intracellular DC-ASGPR was localized to early endosomes, suggesting that the receptor recycles to the cell surface following internalization of ligand.
  • DC-ASGPR is known to be capable of directing the internalization of surrogate antigen into human DC
  • the invention uses novel biological activities of DC-ASGPR to effect particularly desirable changes in the immune system, some in the context of antigen uptake (e.g., vaccination), others through the unique action of DC-ASGPR effectors (alone or in concert with other immune regulatory molecules) capable of eliciting signaling through this receptor on DC, B cells, and monocytes.
  • the invention disclosure reveals means of developing unique agents capable of activating cells bearing DC-ASGPR, as well as the effect of the resulting changes in cells receiving these signals regards action on other cells in the immune system.
  • These effects are highly predictive of therapeutic outcomes for certain disease states or for augmenting protective outcomes in the context of vaccination.
  • the present invention includes compositions and methods for increasing the effectiveness of antigen presentation by a DC-ASGPR-expressing antigen presenting cell by isolating and purifying a DC-ASGPR-specific antibody or fragment thereof to which a targeted agent is attached that forms an antibody-antigen complex, wherein the agent is processed and presented by, e.g., a dendritic cell, that has been contacted with the antibody-agent complex.
  • the antigen presenting cell is a dendritic cell and the DC-ASGPR-specific antibody or fragment thereof is bound to one half of a Coherin/Dockerin pair.
  • the DC-ASGPR-specific antibody or fragment thereof may also be bound to one half of a Coherin/Dockerin pair and an antigen is bound to the complementary half of the Coherin/Dockerin pair to form a complex.
  • agents include one or more peptides, proteins, lipids, carbohydrates, nucleic acids and combinations thereof.
  • the agent may one or more cytokine selected from interleukins, transforming growth factors (TGFs), fibroblast growth factors (FGFs), platelet derived growth factors (PDGFs), epidermal growth factors (EGFs), connective tissue activated peptides (CTAPs), osteogenic factors, and biologically active analogs, fragments, and derivatives of such growth factors, B/T-cell differentiation factors, B/T-cell growth factors, mitogenic cytokines, chemotactic cytokines, colony stimulating factors, angiogenesis factors, IFN- ⁇ , IFN- ⁇ , IFN- ⁇ , IL1, IL2, IL3, IL4, IL5, IL6, IL7, IL8, IL9, IL10, IL11, IL12, IL13, IL14, IL15, IL16, IL17, IL18, etc., leptin, myostatin, macrophage stimulating protein, platelet-derived growth factor, TNF- ⁇ , TNF- ⁇ , NGF
  • the present invention also includes compositions and methods for increasing the effectiveness of antigen presentation by dendritic cells comprising binding a DC-ASGPR-specific antibody or fragment thereof to which an antigen is attached that forms an antibody-antigen complex, wherein the antigen is processed and presented by a dendritic cell that has been contacted with the antibody-antigen complex.
  • Another embodiment is the use of antibodies or other specific binding molecules directed to DC-AS GPR for delivering antigens to antigen-presenting cells for the purpose of eliciting protective or therapeutic immune responses.
  • antigen-targeting reagents specific to DC-ASGPR for vaccination via the skin; antigen-targeting reagents specific to DC-AS GPR in association with co-administered or linked adjuvant for vaccination or use for antigen-targeting (vaccination) purposes of specific antigens which can be expressed as recombinant antigen-antibody fusion proteins.
  • Another embodiment includes a method for increasing the effectiveness of dendritic cells by isolating patient dendritic cells; exposing the dendritic cells to activating amounts of anti-DC-ASGPR antibodies or fragments thereof and antigen to form antigen-loaded, activated dendritic cells; and reintroducing the antigen-loaded, activated dendritic cells into the patient.
  • the antigen may be a bacterial, viral, fungal, protozoan or cancer protein.
  • the present invention also includes an anti-DC-AS GPR immunoglobulin or portion thereof that is secreted from mammalian cells and an antigen bound to the immunoglobulin.
  • the immunoglobulin is bound to one half of a cohesin/dockerin domain, or it may also include a complementary half of the cohesin-dockerin binding pair bound to an antigen that forms a complex with the modular rAb carrier, or a complementary half of the cohesin-dockerin binding pair that is a fusion protein with an antigen.
  • the antigen specific domain may be a full length antibody, an antibody variable region domain, an Fab fragment, a Fab′ fragment, an F(ab) 2 fragment, and Fv fragment, and Fabc fragment and/or a Fab fragment with portions of the Fc domain.
  • the anti-DC-ASGPR immunoglobulin may also be bound to a toxin selected from wherein the toxin is selected from the group consisting of a radioactive isotope, metal, enzyme, botulin, tetanus, ricin, cholera, diphtheria, aflatoxins, perfringens toxin, mycotoxins, shigatoxin, staphylococcal enterotoxin B, T2, seguitoxin, saxitoxin, abrin, cyanoginosin, alphatoxin, tetrodotoxin, aconotoxin, snake venom and spider venom.
  • the antigen may be a fusion protein with the immunoglobulin or bound chemically covalently or not.
  • the present invention also includes compositions and methods for increasing the effectiveness of dendritic cells by isolating patient dendritic cells, exposing the dendritic cells to activating amounts of anti-DC-ASGPR antibodies or fragments thereof and antigen to form antigen-loaded, activated dendritic cells; and reintroducing the antigen-loaded, activated dendritic cells into the patient.
  • the agents may be used to engage DC-ASGPR, alone or with co-activating agents, to activate antigen-presenting cells for therapeutic or protective applications, to bind DC-ASGPR and/or activating agents linked to antigens, alone or with co-activating agents, for protective or therapeutic vaccination.
  • Another use of is the development of specific antibody V-region sequences capable of binding to and activating DC-ASGPR, for use as anti-DC-ASGPR agents linked to toxic agents for therapeutic purposes in the context of diseases known or suspected to result from inappropriate activation of immune cells via DC-ASGPR and as a vaccine with a DC-ASGPR-specific antibody or fragment thereof to which an antigen is attached that forms an antibody-antigen complex, wherein the antigen is processed and presented by a dendritic cell that has been contacted with the antibody-antigen complex.
  • FIGS. 1A to 1E demonstrate signaling through lectin-like receptor DC-AS GPR activates DCs, resulting in increased levels of costimulatory molecules as well as cytokines and chemokines.
  • FIG. 1A shows three day and six day GM/IL-4 DCs were stained with FITC-labeled goat anti-mouse IgG followed by mouse monoclonal anti-human DC-ASGPR, antibody.
  • FIG. 1B shows six day GM/IL-4 DCs were cultured in plates coated with the anti-DC-ASGPR or control mAbs (1-2 ug/ml) for 16-18 h. Cells were stained with anti-CD86 and HLA-DR antibodies labeled with fluorescent dyes. Open and filled bars in the histograms represent cells activated with isotype control mAbs and anti-lectin mAbs, respectively.
  • FIG. 1C shows six day GM/IL-4 DCs were cultured in plates coated with the mAbs for 12 h, and subjected to RNA isolation and Affymetrix Gene Chip analysis, as described in Methods.
  • FIG. 1D shows the cytokines and chemokines in the culture supernatants from the experiment shown in FIG. 1B were measured by Luminex.
  • FIG. 1E shows six day GM/IL-4 DCs were cultured in plates coated with mAbs in the presence or absence of 50 ng/ml soluble CD40L, for 16-18 h, and then stained with anti-CD83 antibodies. Cytokines and chemokines in the culture supernatants from the experiment shown in FIG. 1E were measured by Luminex. Results shown are representative of three independent experiments using cells from different normal donors.
  • FIGS. 2A to 2D shows that DC-ASGPR expressed on DCs, contributes to enhanced humoral immune responses.
  • FIG. 2A is a FACS of day six cells stained with fluorescently labeled antibodies. CD3 + and 7-AAD + cells were gated out. CD38 + and CFSE ⁇ cells were purified by FACS sorter and Giemsa staining was performed.
  • FIG. 1A is a FACS of day six cells stained with fluorescently labeled antibodies.
  • CD3 + and 7-AAD + cells were gated out.
  • CD38 + and CFSE ⁇ cells were purified by FACS sorter and Gi
  • FIG. 1C shows DCs pulsed with 5 multiplicity of infection (moi) of heat-inactivated influenza virus (PR8), and cultured with B cells. Culture supernatant was analyzed for influenza-specific immunoglobulins (Igs) on day thirteen.
  • FIG. 1D shows DC cultured with anti-DC-ASGPR or control mAb were stained for cell surface APRIL expression and the supernatants assayed for soluble APRIL.
  • FIGS. 3A to 3D shows the cell surface expression of DC-ASGPR on B cells contribute to B cell activation and immunoglobulin production.
  • FIG. 3A are PBMCs from buffy coats were stained with anti-CD19, anti-CD3, and anti-DC-ASGPR or control mAb. CD19 + and CD3 + cells were gated and the expression levels of the molecules on CD19 + B cells were measured by flow cytometry.
  • FIG. 3B are CD19 + B cells from buffy coats were cultured in plates coated with the mAbs for 12 h, and subjected to RNA isolation and Affymetrix Gene Chip analysis as described in Methods. Fold increases of gene expression by anti-DC-ASGPR mAb were compared to the gene expression levels in CD19 + B cells stimulated with control mAb.
  • FIG. 3C shows CD19 + B cells were cultured in plates coated with the mAbs for 16-18 h, and then culture supernatants were analyzed for cytokines and chemokines by Luminex.
  • FIG. 3D shows 1 ⁇ 10 5 CD19 + B cells were cultured in plates coated with the mAbs for thirteen days. Total Ig levels were measured by ELISA. Data are representative of two repeat experiments using cells from three different normal donors.
  • FIGS. 4A to 4D shows that the proliferation of purified allogeneic T cells was significantly enhanced by DCs stimulated with mAb specific for DC-ASGPR.
  • FIG. 5 shows that certain anti-DC-ASGPR mAbs can activate DC.
  • DC were incubated for 24 hrs with one of a panel of 12 pure anti-ASGPR mAbs.
  • Cells were then tested for expression of cell surface CD86 (a DC activation marker) and supernatants were assayed for secreted cytokines.
  • Three mAbs (36, 38, 43) from the anti-ASGPR mAb panel activated DC.
  • FIG. 6 shows that different antigens can be expressed in the context of a DC-ASGPR rAb.
  • Such an anti-DC-ASGPR rAb.Doc protein can be simply mixed with any Cohesin.fusion protein to assemble a stable non-covalent [rAb.Doc:Coh.fusion] complex that functions just as a rAb.fusion protein.
  • FIG. 7 GM-CSF/IFNa DCs (5,000/well) were loaded with 10 or 1 nM anti-DC-ASGPR.Doc:Coh.Flu M1, or hIgG4.Doc:Coh.Flu M1 complexes. After 6 h, autologous CD8+ T cells (200,000/well) were added into the cultures. At day 8, the CD8+ T cells were analyzed for expansion of cells bearing TCR specific for a HLA-A201 immuno-dominant peptide. The inner boxes indicate the percentage of tetramer-specific CD8+ T cells.
  • FIGS. 8A-8D demonstrated the cross reactivity of the different antibodies with monkey ASGPR.
  • DCs Dendritic cells
  • Mellman and Steinman 2001 (Banchereau, Briere et al. 2000), (Cella, Sallusto et al. 1997).
  • DCs capture antigens, process them into peptides, and present these to T cells. Therefore delivering antigens directly to DC is a focus area for improving vaccines.
  • One such example is the development of DC-based vaccines using ex-vivo antigen-loading of autologous DCs that are then re-administrated to patients (Banchereau, Schuler-Thurner et al. 2001), (Steinman and Dhodapkar 2001).
  • Another strategy to improve vaccine efficacy is specific targeting to DC of antigen conjugated to antibodies against internalizing DC-specific receptors.
  • the potential of targeting DCfor vaccination is highlighted by key mouse studies.
  • targeting with an anti-LOX-1 mAb coupled to ovalbumin (OVA) induced a protective CD8+ T cell response, via exogenous antigen cross-presentation toward the MHC class I pathway (Delneste, Magistrelli et al. 2002).
  • OVA conjugated to anti-DEC205 mAb in combination with a CD40L maturation stimulus enhanced the MHC class I-restricted presentation by DCs in vivo and led to the durable formation of effector memory CD8+ T cells (Bonifaz, Bonnyay et al. 2004).
  • the present invention provides for the complexing of multiple antigens or proteins (engineered, expressed, and purified independently from the primary mAb) in a controlled, multivariable fashion, to one single primary recombinant mAb.
  • multiple antigens or proteins engineered, expressed, and purified independently from the primary mAb
  • present invention provides for addition to the primary mAb of multiple combinations, in fixed equimolar ratios and locations, of separately engineered proteins.
  • the term “modular rAb carrier” is used to describe a recombinant antibody system that has been engineered to provide the controlled modular addition of diverse antigens, activating proteins, or other antibodies to a single recombinant monoclonal antibody (mAb).
  • the rAb may be a monoclonal antibody made using standard hybridoma techniques, recombinant antibody display, humanized monoclonal antibodies and the like.
  • the modular rAb carrier can be used to, e.g., target (via one primary recombinant antibody against an internalizing receptor, e.g., a human dendritic cell receptor) multiple antigens and/or antigens and an activating cytokine to dendritic cells (DC).
  • the modular rAb carrier may also be used to join two different recombinant mAbs end-to-end in a controlled and defined manner.
  • the antigen binding portion of the “modular rAb carrier” may be one or more variable domains, one or more variable and the first constant domain, an Fab fragment, a Fab′ fragment, an F(ab) 2 fragment, and Fv fragment, and Fabc fragment and/or a Fab fragment with portions of the Fc domain to which the cognate modular binding portions are added to the amino acid sequence and/or bound.
  • the antibody for use in the modular rAb carrier can be of any isotype or class, subclass or from any source (animal and/or recombinant).
  • the modular rAb carrier is engineered to have one or more modular cohesin-dockerin protein domains for making specific and defined protein complexes in the context of engineered recombinant mAbs.
  • the mAb is a portion of a fusion protein that includes one or more modular cohesin-dockerin protein domains carboxy from the antigen binding domains of the mAb.
  • the cohesin-dockerin protein domains may even be attached post-translationally, e.g., by using chemical cross-linkers and/or disulfide bonding.
  • antigen refers to a molecule that can initiate a humoral and/or cellular immune response in a recipient of the antigen.
  • Antigen may be used in two different contexts with the present invention: as a target for the antibody or other antigen recognition domain of the rAb or as the molecule that is carried to and/or into a cell or target by the rAb as part of a dockerin/cohesin-molecule complement to the modular rAb carrier.
  • the antigen is usually an agent that causes a disease for which a vaccination would be advantageous treatment.
  • the peptide is often about 8 to about 25 amino acids.
  • Antigens include any type of biologic molecule, including, for example, simple intermediary metabolites, sugars, lipids and hormones as well as macromolecules such as complex carbohydrates, phospholipids, nucleic acids and proteins.
  • Common categories of antigens include, but are not limited to, viral antigens, bacterial antigens, fungal antigens, protozoal and other parasitic antigens, tumor antigens, antigens involved in autoimmune disease, allergy and graft rejection, and other miscellaneous antigens.
  • the modular rAb carrier is able to carry any number of active agents, e.g., antibiotics, anti-infective agents, antiviral agents, anti-tumoral agents, antipyretics, analgesics, anti-inflammatory agents, therapeutic agents for osteoporosis, enzymes, cytokines, anticoagulants, polysaccharides, collagen, cells, and combinations of two or more of the foregoing active agents.
  • active agents e.g., antibiotics, anti-infective agents, antiviral agents, anti-tumoral agents, antipyretics, analgesics, anti-inflammatory agents, therapeutic agents for osteoporosis, enzymes, cytokines, anticoagulants, polysaccharides, collagen, cells, and combinations of two or more of the foregoing active agents.
  • antibiotics for delivery using the present invention include, without limitation, tetracycline, aminoglycosides, penicillins, cephalosporins, sulfonamide drugs, chloramphenicol sodium succinate, erythromycin, vancomycin, lincomycin, clindamycin, nystatin, amphotericin B, amantidine, idoxuridine, p-amino salicyclic acid, isoniazid, rifampin, antinomycin D, mithramycin, daunomycin, adriamycin, bleomycin, vinblastine, vincristine, procarbazine, imidazole carboxamide, and the like.
  • anti-tumor agents for delivery using the present invention include, without limitation, doxorubicin, Daunorubicin, taxol, methotrexate, and the like.
  • antipyretics and analgesics include aspirin, Motrin®, Ibuprofen®, naprosyn, acetaminophen, and the like.
  • anti-inflammatory agents for delivery using the present invention include, without limitation, include NSAIDS, aspirin, steroids, dexamethasone, hydrocortisone, prednisolone, Diclofenac Na, and the like.
  • therapeutic agents for treating osteoporosis and other factors acting on bone and skeleton include for delivery using the present invention include, without limitation, calcium, alendronate, bone GLa peptide, parathyroid hormone and its active fragments, histone H4-related bone formation and proliferation peptide and mutations, derivatives and analogs thereof.
  • enzymes and enzyme cofactors for delivery using the present invention include, without limitation, pancrease, L-asparaginase, hyaluronidase, chymotrypsin, trypsin, tPA, streptokinase, urokinase, pancreatin, collagenase, trypsinogen, chymotrypsinogen, plasminogen, streptokinase, adenyl cyclase, superoxide dismutase (SOD), and the like.
  • SOD superoxide dismutase
  • cytokines for delivery using the present invention include, without limitation, interleukins, transforming growth factors (TGFs), fibroblast growth factors (FGFs), platelet derived growth factors (PDGFs), epidermal growth factors (EGFs), connective tissue activated peptides (CTAPs), osteogenic factors, and biologically active analogs, fragments, and derivatives of such growth factors.
  • TGFs transforming growth factors
  • FGFs fibroblast growth factors
  • PDGFs platelet derived growth factors
  • EGFs epidermal growth factors
  • CRFs connective tissue activated peptides
  • osteogenic factors and biologically active analogs, fragments, and derivatives of such growth factors.
  • Cytokines may be B/T-cell differentiation factors, B/T-cell growth factors, mitogenic cytokines, chemotactic cytokines, colony stimulating factors, angiogenesis factors, IFN- ⁇ , IFN- ⁇ , IFN- ⁇ , IL1, IL2, IL3, IL4, IL5, IL6, IL7, IL8, IL9, IL10, IL11, IL12, IL13, IL14, IL15, IL16, IL17, IL18, etc., leptin, myostatin, macrophage stimulating protein, platelet-derived growth factor, TNF- ⁇ , TNF- ⁇ , NGF, CD40L, CD137L/4-1BBL, human lymphotoxin- ⁇ , G-CSF, M-CSF, GM-CSF, PDGF, IL-1 ⁇ , IL1- ⁇ , IP-10, PF4, GRO, 9E3, erythropoietin, endostatin, angiostatin, VE
  • TGF transforming growth factor
  • Other cytokines include members of the transforming growth factor (TGF) supergene family include the beta transforming growth factors (for example TGF- ⁇ 1, TGF- ⁇ 2, TGF- ⁇ 3); bone morphogenetic proteins (for example, BMP-1, BMP-2, BMP-3, BMP-4, BMP-5, BMP-6, BMP-7, BMP-8, BMP-9); heparin-binding growth factors (for example, fibroblast growth factor (FGF), epidermal growth factor (EGF), platelet-derived growth factor (PDGF), insulin-like growth factor (IGF)); Inhibins (for example, Inhibin A, Inhibin B); growth differentiating factors (for example, GDF-1); and Activins (for example, Activin A, Activin B, Activin AB).
  • TGF transforming growth factor
  • BMP-3 bone morphogenetic proteins
  • BMP-5, BMP-6, BMP-7, BMP-8, BMP-9 heparin-bind
  • growth factors for delivery using the present invention include, without limitation, growth factors that can be isolated from native or natural sources, such as from mammalian cells, or can be prepared synthetically, such as by recombinant DNA techniques or by various chemical processes.
  • analogs, fragments, or derivatives of these factors can be used, provided that they exhibit at least some of the biological activity of the native molecule.
  • analogs can be prepared by expression of genes altered by site-specific mutagenesis or other genetic engineering techniques.
  • anticoagulants for delivery using the present invention include, without limitation, include warfarin, heparin, Hirudin, and the like.
  • factors acting on the immune system include for delivery using the present invention include, without limitation, factors which control inflammation and malignant neoplasms and factors which attack infective microorganisms, such as chemotactic peptides and bradykinins.
  • viral antigens include, but are not limited to, e.g., retroviral antigens such as retroviral antigens from the human immunodeficiency virus (HIV) antigens such as gene products of the gag, pol, and env genes, the Nef protein, reverse transcriptase, and other HIV components; hepatitis viral antigens such as the S, M, and L proteins of hepatitis B virus, the pre-S antigen of hepatitis B virus, and other hepatitis, e.g., hepatitis A, B, and C, viral components such as hepatitis C viral RNA; influenza viral antigens such as hemagglutinin and neuraminidase and other influenza viral components; measles viral antigens such as the measles virus fusion protein and other measles virus components; rubella viral antigens such as proteins E1 and E2 and other rubella virus components; rotaviral antigens such as VP7
  • Antigenic targets that may be delivered using the rAb-DC/DC-antigen vaccines of the present invention include genes encoding antigens such as viral antigens, bacterial antigens, fungal antigens or parasitic antigens.
  • Viruses include picornavirus, coronavirus, togavirus, flavirvirus, rhabdovirus, paramyxovirus, orthomyxovirus, bunyavirus, arenavirus, reovirus, retrovirus, papilomavirus, parvovirus, herpesvirus, poxvirus, hepadnavirus, and spongiform virus.
  • Other viral targets include influenza, herpes simplex virus 1 and 2, measles, dengue, smallpox, polio or HIV.
  • Pathogens include trypanosomes, tapeworms, roundworms, helminthes, and malaria.
  • Tumor markers such as fetal antigen or prostate specific antigen, may be targeted in this manner.
  • Other examples include: HIV env proteins and hepatitis B surface antigen.
  • Administration of a vector according to the present invention for vaccination purposes would require that the vector-associated antigens be sufficiently non-immunogenic to enable long term expression of the transgene, for which a strong immune response would be desired. In some cases, vaccination of an individual may only be required infrequently, such as yearly or biennially, and provide long term immunologic protection against the infectious agent.
  • Bacterial antigens for use with the rAb vaccine disclosed herein include, but are not limited to, e.g., bacterial antigens such as pertussis toxin, filamentous hemagglutinin, pertactin, FIM2, FIM3, adenylate cyclase and other pertussis bacterial antigen components; diptheria bacterial antigens such as diptheria toxin or toxoid and other diptheria bacterial antigen components; tetanus bacterial antigens such as tetanus toxin or toxoid and other tetanus bacterial antigen components; streptococcal bacterial antigens such as M proteins and other streptococcal bacterial antigen components; gram-negative bacilli bacterial antigens such as lipopolysaccharides and other gram-negative bacterial antigen components, Mycobacterium tuberculosis bacterial antigens such as mycolic acid, heat shock protein
  • Partial or whole pathogens may also be: haemophilus influenza; Plasmodium falciparum; Neisseria meningitidis; Streptococcus pneumoniae; Neisseria gonorrhoeae; salmonella serotype typhi; shigella; Vibrio cholerae ; Dengue Fever; Encephalitides; Japanese Encephalitis; Lyme disease; Yersinia pestis ; west nile virus; yellow fever; tularemia; hepatitis (viral; bacterial); RSV (respiratory syncytial virus); HPIV 1 and HPIV 3; adenovirus; small pox; allergies and cancers.
  • Fungal antigens for use with compositions and methods of the invention include, but are not limited to, e.g., candida fungal antigen components; histoplasma fungal antigens such as heat shock protein 60 (HSP60) and other histoplasma fungal antigen components; cryptococcal fungal antigens such as capsular polysaccharides and other cryptococcal fungal antigen components; coccidiodes fungal antigens such as spherule antigens and other coccidiodes fungal antigen components; and tinea fungal antigens such as trichophytin and other coccidiodes fungal antigen components.
  • candida fungal antigen components histoplasma fungal antigens such as heat shock protein 60 (HSP60) and other histoplasma fungal antigen components
  • cryptococcal fungal antigens such as capsular polysaccharides and other cryptococcal fungal antigen components
  • coccidiodes fungal antigens such as spherule antigen
  • protozoal and other parasitic antigens include, but are not limited to, e.g., Plasmodium falciparum antigens such as merozoite surface antigens, sporozoite surface antigens, circumsporozoite antigens, gametocyte/gamete surface antigens, blood-stage antigen pf 155/RESA and other plasmodial antigen components; toxoplasma antigens such as SAG-1, p30 and other toxoplasmal antigen components; schistosomae antigens such as glutathione-S-transferase, paramyosin, and other schistosomal antigen components; Leishmania major and other leishmaniae antigens such as gp63, lipophosphoglycan and its associated protein and other leishmanial antigen components; and Trypanosoma cruzi antigens such as the 75-77 kDa antigen, the 56 kDa antigen and other trypanosomal antigen
  • Antigen that can be targeted using the rAb of the present invention will generally be selected based on a number of factors, including: likelihood of internalization, level of immune cell specificity, type of immune cell targeted, level of immune cell maturity and/or activation and the like.
  • cell surface markers for dendritic cells include, but are not limited to, MHC class I, MHC Class II, B7-2, CD18, CD29, CD31, CD43, CD44, CD45, CD54, CD58, CD83, CD86, CMRF-44, CMRF-56, DCIR and/or ASPGR and the like; while in some cases also having the absence of CD2, CD3, CD4, CD8, CD14, CD15, CD16, CD 19, CD20, CD56, and/or CD57.
  • cell surface markers for antigen presenting cells include, but are not limited to, MHC class I, MHC Class II, CD40, CD45, B7-1, B7-2, IFN- ⁇ receptor and IL-2 receptor, ICAM-1 and/or Fc ⁇ receptor.
  • cell surface markers for T cells include, but are not limited to, CD3, CD4, CD8, CD 14, CD20, CD11b, CD16, CD45 and HLA-DR.
  • Target antigens on cell surfaces for delivery includes those characteristic of tumor antigens typically will be derived from the cell surface, cytoplasm, nucleus, organelles and the like of cells of tumor tissue.
  • tumor targets for the antibody portion of the present invention include, without limitation, hematological cancers such as leukemias and lymphomas, neurological tumors such as astrocytomas or glioblastomas, melanoma, breast cancer, lung cancer, head and neck cancer, gastrointestinal tumors such as gastric or colon cancer, liver cancer, pancreatic cancer, genitourinary tumors such cervix, uterus, ovarian cancer, vaginal cancer, testicular cancer, prostate cancer or penile cancer, bone tumors, vascular tumors, or cancers of the lip, nasopharynx, pharynx and oral cavity, esophagus, rectum, gall bladder, biliary tree, larynx, lung and bronchus, bladder, kidney, brain and other parts of the nervous system,
  • antigens examples include tumor proteins, e.g., mutated oncogenes; viral proteins associated with tumors; and tumor mucins and glycolipids.
  • the antigens may be viral proteins associated with tumors would be those from the classes of viruses noted above.
  • Certain antigens may be characteristic of tumors (one subset being proteins not usually expressed by a tumor precursor cell), or may be a protein which is normally expressed in a tumor precursor cell, but having a mutation characteristic of a tumor.
  • Other antigens include mutant variant(s) of the normal protein having an altered activity or subcellular distribution, e.g., mutations of genes giving rise to tumor antigens.
  • tumor antigens include: CEA, prostate specific antigen (PSA), HER-2/neu, BAGE, GAGE, MAGE 1-4, 6 and 12, MUC (Mucin) (e.g., MUC-1, MUC-2, etc.), GM2 and GD2 gangliosides, ras, myc, tyrosinase, MART (melanoma antigen), Pmel 17(gp100), GnT-V intron V sequence (N-acetylglucoaminyltransferase V intron V sequence), Prostate Ca psm, PRAME (melanoma antigen), ⁇ -catenin, MUM-1-B (melanoma ubiquitous mutated gene product), GAGE (melanoma antigen) 1, BAGE (melanoma antigen) 2-10, c-ERB2 (Her2/neu), EBNA (Epstein-Barr Virus nuclear antigen) 1-6, gp75, human papillo
  • the immunogenic molecule can be an autoantigen involved in the initiation and/or propagation of an autoimmune disease, the pathology of which is largely due to the activity of antibodies specific for a molecule expressed by the relevant target organ, tissue, or cells, e.g., SLE or MG.
  • a Th2-type immune response to the relevant autoantigen towards a cellular (i.e., a Th1-type) immune response.
  • Autoantigens of interest include, without limitation: (a) with respect to SLE, the Smith protein, RNP ribonucleoprotein, and the SS-A and SS-B proteins; and (b) with respect to MG, the acetylcholine receptor.
  • examples of other miscellaneous antigens involved in one or more types of autoimmune response include, e.g., endogenous hormones such as luteinizing hormone, follicular stimulating hormone, testosterone, growth hormone, prolactin, and other hormones.
  • Antigens involved in autoimmune diseases, allergy, and graft rejection can be used in the compositions and methods of the invention.
  • an antigen involved in any one or more of the following autoimmune diseases or disorders can be used in the present invention: diabetes, diabetes mellitus, arthritis (including rheumatoid arthritis, juvenile rheumatoid arthritis, osteoarthritis, psoriatic arthritis), multiple sclerosis, myasthenia gravis, systemic lupus erythematosis, autoimmune thyroiditis, dermatitis (including atopic dermatitis and eczematous dermatitis), psoriasis, Sjogren's Syndrome, including keratoconjunctivitis sicca secondary to Sjogren's Syndrome, alopecia areata, allergic responses due to arthropod bite reactions, Crohn's disease, aphthous ulcer, ulceris, conjunctivitis, keratoconjunctivitis, ulcerative colitis, asthma
  • antigens involved in autoimmune disease include glutamic acid decarboxylase 65 (GAD 65), native DNA, myelin basic protein, myelin proteolipid protein, acetylcholine receptor components, thyroglobulin, and the thyroid stimulating hormone (TSH) receptor.
  • GID 65 glutamic acid decarboxylase 65
  • native DNA myelin basic protein
  • myelin proteolipid protein acetylcholine receptor components
  • thyroglobulin thyroid stimulating hormone
  • antigens involved in allergy include pollen antigens such as Japanese cedar pollen antigens, ragweed pollen antigens, rye grass pollen antigens, animal derived antigens such as dust mite antigens and feline antigens, histocompatiblity antigens, and penicillin and other therapeutic drugs.
  • antigens involved in graft rejection include antigenic components of the graft to be transplanted into the graft recipient such as heart, lung, liver, pancreas, kidney, and neural graft components.
  • the antigen may be an altered peptide ligand useful in treating an autoimmune disease.
  • epitope(s) refer to a peptide or protein antigen that includes a primary, secondary or tertiary structure similar to an epitope located within any of a number of pathogen polypeptides encoded by the pathogen DNA or RNA.
  • the level of similarity will generally be to such a degree that monoclonal or polyclonal antibodies directed against such polypeptides will also bind to, react with, or otherwise recognize, the peptide or protein antigen.
  • Various immunoassay methods may be employed in conjunction with such antibodies, such as, for example, Western blotting, ELISA, RIA, and the like, all of which are known to those of skill in the art.
  • pathogen epitopes and/or their functional equivalents, suitable for use in vaccines is part of the present invention. Once isolated and identified, one may readily obtain functional equivalents. For example, one may employ the methods of Hopp, as taught in U.S. Pat. No. 4,554,101, incorporated herein by reference, which teaches the identification and preparation of epitopes from amino acid sequences on the basis of hydrophilicity. The methods described in several other papers, and software programs based thereon, can also be used to identify epitopic core sequences (see, for example, Jameson and Wolf, 1988; Wolf et al., 1988; U.S. Pat. No. 4,554,101). The amino acid sequence of these “epitopic core sequences” may then be readily incorporated into peptides, either through the application of peptide synthesis or recombinant technology.
  • the preparation of vaccine compositions that includes the nucleic acids that encode antigens of the invention as the active ingredient, may be prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid prior to infection can also be prepared.
  • the preparation may be emulsified, encapsulated in liposomes.
  • the active immunogenic ingredients are often mixed with carriers which are pharmaceutically acceptable and compatible with the active ingredient.
  • pharmaceutically acceptable carrier refers to a carrier that does not cause an allergic reaction or other untoward effect in subjects to whom it is administered.
  • suitable pharmaceutically acceptable carriers include, for example, one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol, or the like and combinations thereof.
  • the vaccine can contain minor amounts of auxiliary substances such as wetting or emulsifying agents, pH buffering agents, and/or adjuvants which enhance the effectiveness of the vaccine.
  • adjuvants examples include but are not limited to: aluminum hydroxide, N-acetyl-muramyl-L-threonyl-D-isoglutamine (thr-MDP), N-acetyl-nor-muramyl-L-alanyl-D-isoglutamine, MTP-PE and RIB I, which contains three components extracted from bacteria, monophosporyl lipid A, trehalose dimycolate and cell wall skeleton (MPL+TDM+CWS) in a 2% squalene/Tween 80 emulsion.
  • thr-MDP N-acetyl-muramyl-L-threonyl-D-isoglutamine
  • MTP-PE N-acetyl-nor-muramyl-L-alanyl-D-isoglutamine
  • MTP-PE N-acetyl-nor-muramyl-L-alanyl-D-isoglutamine
  • adjuvants include DDA (dimethyldioctadecylammonium bromide), Freund's complete and incomplete adjuvants and QuilA.
  • immune modulating substances such as lymphokines (e.g., IFN- ⁇ , IL-2 and IL-12) or synthetic IFN- ⁇ inducers such as poly I:C can be used in combination with adjuvants described herein.
  • compositions may include a naked polynucleotide with a single or multiple copies of the specific nucleotide sequences that bind to specific DNA-binding sites of the apolipoproteins present on plasma lipoproteins as described in the current invention.
  • the polynucleotide may encode a biologically active peptide, antisense RNA, or ribozyme and will be provided in a physiologically acceptable administrable form.
  • Another pharmaceutical product that may spring from the current invention may include a highly purified plasma lipoprotein fraction, isolated according to the methodology, described herein from either the patients blood or other source, and a polynucleotide containing single or multiple copies of the specific nucleotide sequences that bind to specific DNA-binding sites of the apolipoproteins present on plasma lipoproteins, prebound to the purified lipoprotein fraction in a physiologically acceptable, administrable form.
  • Yet another pharmaceutical product may include a highly purified plasma lipoprotein fraction which contains recombinant apolipoprotein fragments containing single or multiple copies of specific DNA-binding motifs, prebound to a polynucleotide containing single or multiple copies of the specific nucleotide sequences, in a physiologically acceptable administrable form.
  • Yet another pharmaceutical product may include a highly purified plasma lipoprotein fraction which contains recombinant apolipoprotein fragments containing single or multiple copies of specific DNA-binding motifs, prebound to a polynucleotide containing single or multiple copies of the specific nucleotide sequences, in a physiologically acceptable administrable form.
  • the dosage to be administered depends to a great extent on the body weight and physical condition of the subject being treated as well as the route of administration and frequency of treatment.
  • a pharmaceutical composition that includes the naked polynucleotide prebound to a highly purified lipoprotein fraction may be administered in amounts ranging from 1 ⁇ g to 1 mg polynucleotide and 1 ⁇ g to 100 mg protein.
  • rAb and rAb complexes a patient will follow general protocols for the administration of chemotherapeutics, taking into account the toxicity, if any, of the vector. It is anticipated that the treatment cycles would be repeated as necessary. It also is contemplated that various standard therapies, as well as surgical intervention, may be applied in combination with the described gene therapy.
  • Aqueous compositions of the present invention may include an effective amount of the compound, dissolved or dispersed in a pharmaceutically acceptable carrier or aqueous medium. Such compositions can also be referred to as inocula.
  • a pharmaceutically acceptable carrier or aqueous medium Such compositions can also be referred to as inocula.
  • the use of such media and agents for pharmaceutical active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients also can be incorporated into the compositions.
  • the compositions of the present invention may include classic pharmaceutical preparations. Dispersions also can be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • compositions according to the present invention will be via any common route so long as the target tissue is available via that route in order to maximize the delivery of antigen to a site for maximum (or in some cases minimum) immune response.
  • Administration will generally be by orthotopic, intradermal, subcutaneous, intramuscular, intraperitoneal or intravenous injection. Other areas for delivery include: oral, nasal, buccal, rectal, vaginal or topical. Topical administration would be particularly advantageous for treatment of skin cancers.
  • Such compositions would normally be administered as pharmaceutically acceptable compositions that include physiologically acceptable carriers, buffers or other excipients.
  • Vaccine or treatment compositions of the invention may be administered parenterally, by injection, for example, either subcutaneously or intramuscularly. Additional formulations which are suitable for other modes of administration include suppositories, and in some cases, oral formulations or formulations suitable for distribution as aerosols. In the case of the oral formulations, the manipulation of T-cell subsets employing adjuvants, antigen packaging, or the addition of individual cytokines to various formulation that result in improved oral vaccines with optimized immune responses.
  • binders and carriers may include, for example, polyalkylene glycols or triglycerides; such suppositories may be formed from mixtures containing the active ingredient in the range of 0.5% to 10%, preferably 1%-2%.
  • Oral formulations include such normally employed excipients as, for example, pharmaceutical grades of mannitol, lactose, starch magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, and the like. These compositions take the form of solutions, suspensions, tablets, pills, capsules, sustained release formulations or powders and contain 10%-95% of active ingredient, preferably 25-70%.
  • the antigen encoding nucleic acids of the invention may be formulated into the vaccine or treatment compositions as neutral or salt forms.
  • Pharmaceutically acceptable salts include the acid addition salts (formed with free amino groups of the peptide) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or with organic acids such as acetic, oxalic, tartaric, maleic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine, and the like.
  • Vaccine or treatment compositions are administered in a manner compatible with the dosage formulation, and in such amount as will be prophylactically and/or therapeutically effective.
  • the quantity to be administered depends on the subject to be treated, including, e.g., capacity of the subject's immune system to synthesize antibodies, and the degree of protection or treatment desired.
  • Suitable dosage ranges are of the order of several hundred micrograms active ingredient per vaccination with a range from about 0.1 mg to 1000 mg, such as in the range from about 1 mg to 300 mg, and preferably in the range from about 10 mg to 50 mg.
  • Suitable regiments for initial administration and booster shots are also variable but are typified by an initial administration followed by subsequent inoculations or other administrations.
  • nucleic acid molecule or fusion polypeptides of this invention will depend, inter alia, upon the administration schedule, the unit dose of antigen administered, whether the nucleic acid molecule or fusion polypeptide is administered in combination with other therapeutic agents, the immune status and health of the recipient, and the therapeutic activity of the particular nucleic acid molecule or fusion polypeptide.
  • compositions can be given in a single dose schedule or in a multiple dose schedule.
  • a multiple dose schedule is one in which a primary course of vaccination may include, e.g., 1-10 separate doses, followed by other doses given at subsequent time intervals required to maintain and or reinforce the immune response, for example, at 1-4 months for a second dose, and if needed, a subsequent dose(s) after several months.
  • Periodic boosters at intervals of 1-5 years, usually 3 years, are desirable to maintain the desired levels of protective immunity.
  • the course of the immunization can be followed by in vitro proliferation assays of peripheral blood lymphocytes (PBLs) co-cultured with ESAT6 or ST-CF, and by measuring the levels of IFN- ⁇ released from the primed lymphocytes.
  • PBLs peripheral blood lymphocytes
  • the assays may be performed using conventional labels, such as radionucleotides, enzymes, fluorescent labels and the like. These techniques are known to one skilled in the art and can be found in U.S. Pat. Nos. 3,791,932, 4,174,384 and 3,949,064, relevant portions incorporated by reference.
  • the modular rAb carrier and/or conjugated rAb carrier-(cohesion/dockerin and/or dockerin-cohesin)-antigen complex may be provided in one or more “unit doses” depending on whether the nucleic acid vectors are used, the final purified proteins, or the final vaccine form is used.
  • Unit dose is defined as containing a predetermined-quantity of the therapeutic composition calculated to produce the desired responses in association with its administration, i.e., the appropriate route and treatment regimen.
  • the quantity to be administered, and the particular route and formulation, are within the skill of those in the clinical arts.
  • the subject to be treated may also be evaluated, in particular, the state of the subject's immune system and the protection desired.
  • a unit dose need not be administered as a single injection but may include continuous infusion over a set period of time.
  • Unit dose of the present invention may conveniently may be described in terms of DNA/kg (or protein/Kg) body weight, with ranges between about 0.05, 0.10, 0.15, 0.20, 0.25, 0.5, 1, 10, 50, 100, 1,000 or more mg/DNA or protein/kg body weight are administered.
  • the amount of rAb-DC/DC-antigen vaccine delivered can vary from about 0.2 to about 8.0 mg/kg body weight.
  • 0.4 mg, 0.5 mg, 0.8 mg, 1.0 mg, 1.5 mg, 2.0 mg, 2.5 mg, 3.0 mg, 4.0 mg, 5.0 mg, 5.5 mg, 6.0 mg, 6.5 mg, 7.0 mg and 7.5 mg of the vaccine may be delivered to an individual in vivo.
  • the dosage of rAb-DC/DC-antigen vaccine to be administered depends to a great extent on the weight and physical condition of the subject being treated as well as the route of administration and the frequency of treatment.
  • a pharmaceutical composition that includes a naked polynucleotide prebound to a liposomal or viral delivery vector may be administered in amounts ranging from 1 ⁇ g to 1 mg polynucleotide to 1 ⁇ g to 100 mg protein.
  • compositions may include between about 1 ⁇ g, 5 ⁇ g, 10 ⁇ g, 20 ⁇ g, 30 ⁇ g, 40 ⁇ g, 50 ⁇ g, 60 ⁇ g, 70 ⁇ g, 80 ⁇ g, 100 ⁇ g, 150 ⁇ g, 200 ⁇ g, 250 ⁇ g, 500 ⁇ g, 600 ⁇ g, 700 ⁇ g, 800 ⁇ g, 900 ⁇ g or 1,000 ⁇ g polynucleotide or protein that is bound independently to 1 ⁇ g, 5 ⁇ g, 10 ⁇ g, 20 ⁇ g, 3.0 ⁇ g, 40 ⁇ g 50 ⁇ g, 60 ⁇ g, 70 ⁇ g, 80 ⁇ g, 100 ⁇ g, 150 ⁇ g, 200 ⁇ g, 250 ⁇ g, 500 ⁇ g, 600 ⁇ g, 700 ⁇ g, 800 ⁇ g, 900 ⁇ g, 1 mg, 1.5 mg, 5 mg, 10 mg, 20 mg, 30 mg, 40 mg, 50 mg, 60 mg, 70 mg, 80 mg, 90 mg or
  • the present invention was tested in an in vitro cellular system that measures immune stimulation of human Flu-specific T cells by dendritic cells to which Flu antigen has been targeted.
  • the results shown herein demonstrate the specific expansion of such antigen specific cells at doses of the antigen which are by themselves ineffective in this system.
  • the present invention may also be used to make a modular rAb carrier that is, e.g., a recombinant humanized mAb (directed to a specific human dendritic cell receptor) complexed with protective antigens from Ricin, Anthrax toxin, and Staphylococcus B enterotoxin.
  • a modular rAb carrier that is, e.g., a recombinant humanized mAb (directed to a specific human dendritic cell receptor) complexed with protective antigens from Ricin, Anthrax toxin, and Staphylococcus B enterotoxin.
  • the potential market for this entity is vaccination of all military personnel and stored vaccine held in reserve to administer to large population centers in response to any biothreat related to these agents.
  • the invention has broad application to the design of vaccines in general, both for human and animal use. Industries of interest include the pharmaceutical and biotechnology industries.
  • the present invention includes compositions and methods, including vaccines, that specifically target (deliver) antigens to antigen-presenting cells (APCs) for the purpose of eliciting potent and broad immune responses directed against the antigen.
  • APCs antigen-presenting cells
  • These compositions evoke protective or therapeutic immune responses against the agent (pathogen or cancer) from which the antigen was derived.
  • the invention creates agents that are directly, or in concert with other agents, therapeutic through their specific engagement of a receptor called DC-ASGPR that is expressed on antigen-presenting cells.
  • the novel recombinant humanized mAb (directed to the specific human dendritic cell receptor DC-ASGPR) fused through the antibody (Ab) heavy chain to antigens known or suspected to encode protective antigens.
  • antigens known or suspected to encode protective antigens.
  • These include as examples for vaccination against various agents—hemagglutinins from Influenza H5N1; HIV gag from attenuated toxins from Ricin, Anthrax toxin, and Staphylococcus B enterotoxin; ‘strings’ of antigenic peptides from melanona antigens, etc.
  • the present invention may be used as a preventative or therapeutic vaccination for at risk or infected patients.
  • the invention has broad application for vaccination against many diseases and cancers, both for human and animal use. Industries that can use the present invention include the pharmaceutical and biotechnological.
  • the present invention can be used to target antigens to APC for vaccination purposes. It is not known which antigen internalizing receptor will be best suited for this purpose.
  • the invention describes particularly advantageous features of DC-ASGPR as for this purpose. Furthermore, the invention shows that engaging DC-ASGPR can be beneficial in the sense of activating the immune system with highly predicted significant therapeutic benefit.
  • the present invention includes the development of high affinity monoclonal antibodies against human DC-ASGPR.
  • Receptor ectodomain.hIgG human IgG1Fc
  • AP human placental alkaline phosphatase
  • An expression vector for hDCIR ectodomain.AP was generated using PCR to amplify AP resides 133-1581 (gbIBC0096471) while adding a proximal in-frame Xho I site and a distal TGA stop codon and Not I site. This Xho I—Not I fragment replaced the IgG coding sequence in the above hDCIR ectodomain.IgG vector.
  • DC-ASGPR ectodomain constructs in the same Ig and AP vector series contained inserts encoding (bp 484-1251, gi153832017).
  • DC-ASGPR fusion proteins were produced using the FreeStyleTM 293 Expression System (Invitrogen) according to the manufacturer's protocol (1 mg total plasmid DNA with 1.3 ml 293 Fectin reagent/L of transfection).
  • rAb production equal amounts of vector encoding the H and L chain were co-transfected.
  • Transfected cells are cultured for 3 days, the culture supernatant was harvested and fresh media added with continued incubation for two days. The pooled supernatants were clarified by filtration.
  • Receptor ectodomain.hIgG was purified by HiTrap protein A affinity chromatography with elution by 0.1 M glycine pH 2.7 and then dialyzed versus PBS.
  • mice were immunized intraperitonealy with 20 ⁇ g of receptor ectodomain.hIgGFc fusion protein with Ribi adjuvant, then boosts with 20 ⁇ g antigen 10 days and 15 days later. After 3 months, the mice were boosted again three days prior to taking the spleens. Alternately, mice were injected in the footpad with 1-10 ⁇ g antigen in Ribi adjuvant every 3-4 days over a 30-40 day period. 3-4 days after a final boost, draining lymph nodes were harvested.
  • B cells from spleen or lymph node cells were fused with SP2/O-Ag 14 cells (Shulman, Wilde et al. 1978) using conventional techniques.
  • ELISA was used to screen hybridoma supernatants against the receptor ectodomain fusion protein compared to the fusion partner alone, or versus the receptor ectodomain fused to AP (Bates, Fournier et al. 1999). Positive wells were then screened in FACS using 293F cells transiently transfected with expression plasmids encoding full-length receptor cDNAs. Selected hybridomas were single cell cloned and expanded in CELLine flasks (Intergra).
  • Hybridoma supernatants were mixed with an equal volume of 1.5 M glycine, 3 M NaCl, 1 ⁇ PBS, pH 7.8 and tumbled with MabSelect resin. The resin was washed with binding buffer and eluted with 0.1 M glycine, pH 2.7. Following neutralization with 2 M Tris, mAbs were dialyzed versus PBS.
  • FIGS. 1A to 1D shows that signaling through DC-ASGPR activates DCs.
  • DCs are the primary immune cells that determine the results of immune responses, either induction or tolerance, depending on their activation (15).
  • the role of LLRs in DC activation is not clear yet. Therefore, we tested whether triggering the LLR DC-ASGPR can result in the activation of DCs.
  • Both three and six day in vitro cultured GM/IL-4 DCs express LOX-1, ASGPR, and CLEC-6 ( FIG. 1A ).
  • Six day DCs were stimulated with mAb specific to DC-ASGPR, and data in FIG. 1B show that signals through DC-ASGPR could activate DCs, resulting in the increased expression of CD86 and HLA-DR.
  • DC-ASGPR Triggering DC-ASGPR on DCs also resulted in the increased production of IL-6, MCP-1, IL-12p40, and IL-8 from DCs ( FIG. 1C ).
  • DCs stimulated with DC-ASGPR specific mAb expressed increased levels of multiple genes, including co-stimulatory molecules as well as chemokine and cytokine-related genes ( FIG. 1D ).
  • DCs stimulated through DC-ASGPR induce potent humoral immune responses.
  • DCs play an important role in humoral immune responses by providing signals for both T-dependent and T-independent B cell responses (19-22) and by transferring antigens to B cells (23, 24).
  • signaling through TLR9 as a third signal is necessary for efficient B cell responses (25, 26).
  • DC-ASGPR in DCs-mediated humoral immune responses in the presence of TLR9 ligand, CpG.
  • Six day GM/IL-4 DCs were stimulated with anti-DC-ASGPR mAb, and then purified B cells were co-cultured.
  • DCs activated with anti-DC-ASGPR mAb resulted in remarkably enhanced B cell proliferation (CFSE dilution) and plasma cell differentiation (CD38 + CD20 ⁇ ), compared to DCs stimulated with control mAb.
  • CD38 + CD20 ⁇ B cells have a typical morphology of plasma cells, but they do not express CD138.
  • DC-ASGPR-mediated DC activation contributes to both total and antigen specific humoral immune responses.
  • APCs ex vivo antigen presenting cells
  • APCs stimulated through DC-ASGPR resulted in enhanced B cell proliferation and plasma cell differentiation in the absence (upper panels in FIG. 2 d ) or presence (lower panels in FIG. 2D ) of TLR9 ligand.
  • the total IgM, IgG, and IgA were also significantly increased when PBMCs were cultured in the plates coated with mAb against DC-ASGPR ( FIG. 2 e ).
  • DCs activated by signaling through DC-ASGPR have matured phenotypes and produce large amounts of inflammatory cytokines and chemokines, and both matured DC phenotypes and soluble factors from DCs could contribute to the enhanced B cells responses ( FIG. 2 ).
  • DC-derived B lymphocyte stimulator protein (BLyS, BAFF) and a proliferation-inducing ligand (APRIL) are also important molecules by which DCs can directly regulate human B cell proliferation and function (27-30). Therefore, we tested whether signals through DC-ASGPR could alter the expression levels of BLyS and APRIL.
  • Data in FIG. 2 d show that DCs stimulated through DC-ASGPR expressed increased levels of intracellular APRIL as well as APRIL secreted, but not BLyS (not shown). Expression levels of BLyS and APRIL receptors on B cells in the mixed cultures were measured, but there was no significant change (not shown).
  • DC-ASGPR contributes to B cell activation and Ig production.
  • CD19 + B cells express DC-ASGPR ( FIG. 3A ). Therefore, we tested the role of DC-ASGPR in B cell activation.
  • Data in FIG. 3B show that B cells stimulated through DC-ASGPR produced significantly higher amounts of chemokines.
  • IL-8 and MIP-1a slight increases in IL-6 and TNFa were also observed when B cells were stimulated with the anti-DC-ASGPR mAb, compared to control mAb.
  • Genes related to cell activation were also up-regulated ( FIG. 3C ).
  • B cells produced IgM, IgG, and IgA when they were stimulated through DC-ASGPR ( FIG. 3D ), suggesting that DC-ASGPR could play an important role in the maintenance of normal immunoglobulin levels in vivo.
  • signaling through DC-ASGPR alone did not induce significant B cell proliferation.
  • DCs stimulated through DC-ASGPR express enhanced levels of co-stimulatory molecules and produce increased amounts of cytokines and chemokines (see FIG. 1 ), suggesting that DC-ASGPR contributes to cellular immune responses as well as humoral immune responses. This was tested by a mixed lymphocyte reaction (MLR). Proliferation of purified allogeneic T cells was significantly enhanced by DCs stimulated with mAb specific for DC-ASGPR ( FIG. 4A ). DCs activated through DC-ASGPR could also prime Mart-1-specific CD8 T cells more efficiently than DC stimulated with control mAb (upper panels in FIG. 4B ).
  • DC-ASGPR plays an important role in enhancing DC function, resulting in better priming and cross-priming of antigens to CD8 T cells.
  • the role of DC-ASGPR expressed on the mixture of APCs in PBMCs in activation of T cell responses is shown in FIG. 4C where PBMCs stimulated with mAb to DC-ASGPR resulted in an increased frequency of Flu M1 tetramer specific CD8 T cells compared to DCs stimulated with control mAb.
  • FIG. 4D shows that DCs stimulated through DC-ASGPR significantly increase CD4 T cell proliferation.
  • Antibodies and tetramers Antibodies (Abs) for surface staining of DCs and B cells, including isotype control Abs, were purchased from BD Biosciences (CA). Abs for ELISA were purchased from Bethyl (TX). Anti-BLyS and anti-APRIL were from PeproTech (NJ). Tetramers, HLA-A*0201—GILGFVFTL (SEQ ID NO.: 1) (Flu M1) and HLA-A*0201-ELAGIGILTV (SEQ ID NO.: 2) (Mart-1), were purchased from Beckman Coulter (CA).
  • PBMCs Mononuclear cells and cultures—Monocytes (1 ⁇ 10 6 /ml) from normal donors were cultured in Cellgenics (France) media containing GM-CSF (100 ng/ml) and IL-4 (50 ng/ml) (R&D, CA). For day three and day six, DCs, the same amounts of cytokines were supplemented into the media on day one and day three, respectively.
  • B cells were purified with a negative isolation kit (BD).
  • CD4 and CD8 T cells were purified with magnetic beads coated with anti-CD4 or CD8 (Milteniy, Calif.).
  • PBMCs were isolated from Buffy coats using PercollTM gradients (GE Healthcare UK Ltd, Buckinghamshire, UK) by density gradient centrifugation.
  • DC activation 1 ⁇ 10 5 DCs were cultured in the mAb-coated 96-well plate for 16-18 h. mAbs (1-2 ⁇ g/well) in carbonate buffer, pH 9.4, were incubated for at least 3 h at 37° C. Culture supernatants were harvested and cytokines/chemokines were measured by Luminex (Biorad, CA). For gene analysis, DCs were cultured in the plates coated with mAbs for 8 h. In some experiments, soluble 50 ng/ml of CD40L (R&D, CA) or 50 nM CpG (InVivogen, CA) was added into the cultures.
  • DCs and B cell co-cultures 5 ⁇ 10 3 DCs resuspended in RPMI 1640 with 10% FCS and antibiotics (Biosource, CA) were first cultured in the plates coated with mAbs for at least 6 h, and then 1 ⁇ 10 5 purified autologous B cells labeled with CFSE (Molecular Probes, OR) were added.
  • DCs were pulsed with 5 moi (multiplicity of infection) of heat-inactivated influenza virus (A/PR/8 H1N1) for 2 h, and then mixed with B cells.
  • A/PR/8 H1N1 heat-inactivated influenza virus
  • 5 ⁇ 10 3 DCs were cultured with 1 ⁇ 10 5 purified autologous CD8 T cells or mixed allogeneic T cells.
  • Allogeneic T cells were pulsed with 1 ⁇ Ci/well 3 [H]-thymidine for the final 18 h of incubation, and then cpm were measured by a ⁇ -counter (Wallac, Minn.). 5 ⁇ 10 5 PBMCs/well were cultured in the plates coated with mAbs. The frequency of Mart-1 and Flu M1 specific CD8 T cells was measured by staining cells with anti-CD8 and tetramers on day ten and day seven of the cultures, respectively. 10 ⁇ M of Mart-1 peptide (ELAGIGILTV) (SEQ ID NO.: 2) and 20 nM of recombinant protein containing Mart-1 peptides (see below) were added to the DC and CD8 T cell cultures. 20 nM purified recombinant Flu M1 protein (see below) was add to the PBMC cultures.
  • Monoclonal antibodies were generated by conventional technology. Briefly, six-week-old BALB/c mice were immunized i.p. with 20 ⁇ g of receptor ectodomain.hIgGFc fusion protein with Ribi adjuvant, then boosts with 20 ⁇ g antigen ten days and fifteen days later. After three months, the mice were boosted again three days prior to taking the spleens. Alternately, mice were injected in the footpad with 1-10 ⁇ g antigen in Ribi adjuvant every three to four days over a thirty to forty day period. Three to four days after a final boost, draining lymph nodes were harvested.
  • B cells from spleen or lymph node cells were fused with SP2/0-Ag 14 cells.
  • Hybridoma supernatants were screened to analyze Abs to the receptor ectodomain fusion protein compared to the fusion partner alone, or the receptor ectodomain fused to alkaline phosphatase (44). Positive wells were then screened in FACS using 293F cells transiently transfected with expression plasmids encoding full-length receptor cDNAs. Selected hybridomas were single cell cloned and expanded in CELLine flasks (Integra, CA).
  • Hybridoma supernatants were mixed with an equal volume of 1.5 M glycine, 3 M NaCl, 1 ⁇ PBS, pH 7.8 and tumbled with MabSelect resin. The resin was washed with binding buffer and eluted with 0.1 M glycine, pH 2.7. Following neutralization with 2 M Tris, mAbs were dialyzed versus PBS.
  • ELISA sandwich ELISA was performed to measure total IgM, IgG, and IgA as well as flu-specific immunoglobulins (Igs).
  • Standard human serum (Bethyl) containing known amounts of Igs and human AB serum were used as standard for total Igs and flu-specific Igs, respectively. Flu specific Ab titers, units, in samples were defined as dilution factor of AB serum that shows an identical optical density.
  • the amounts of BAFF and BLyS were measured by ELISA kits (Bender MedSystem, CA).
  • RNA purification and gene analysis Total RNA extracted with RNeasy columns (Qiagen), and analyzed with the 2100 Bioanalyser (Agilent).
  • Biotin-labeled cRNA targets were prepared using the Illumina totalprep labeling kit (Ambion) and hybridized to Sentrix Human6 BeadChips (46K transcripts). These microarrays consist of 50 mer oligonucleotide probes attached to 3 ⁇ m beads which are lodged into microwells etched at the surface of a silicon wafer. After staining with Streptavidin-Cy3, the array surface is imaged using a sub-micron resolution scanner manufactured by Illumina (Beadstation 500X). A gene expression analysis software program, GeneSpring, Version 7.1 (Agilent), was used to perform data analysis.
  • thermocellum (unpublished) inserted between the Nco I and Nhe I sites expressed Coh.His.
  • E. coli strain BL21 (DE3) (Novagen) or T7 Express (NEB), grown in LB at 37° C. with selection for kanamycin resistance (40 ⁇ g/ml) and shaking at 200 rounds/min to mid log phase growth when 120 mg/L IPTG was added. After three hours, the cells were harvested by centrifugation and stored at ⁇ 80° C. E. coli cells from each 1 L fermentation were resuspended in 30 ml ice-cold 50 mM Tris, 1 mM EDTA pH 8.0 (buffer B) with 0.1 ml of protease inhibitor Cocktail II (Calbiochem, CA).
  • the cells were sonicated on ice 2 ⁇ 5 min at setting 18 (Fisher Sonic Dismembrator 60) with a 5 min rest period and then spun at 17,000 r.p.m. (Sorvall SA-600) for 20 min at 4° C.
  • His.Flu M1 purification the 50 ml cell lysate supernatant fraction was passed through 5 ml Q Sepharose beads and 6.25 ml 160 mM Tris, 40 mM imidazole, 4 M NaCl pH 7.9 was added to the Q Sepharose flow through. This was loaded at 4 ml/min onto a 5 ml HiTrap chelating HP column charged with Ni++.
  • the column-bound protein was washed with 20 mM NaPO 4 , 300 mM NaCl pH 7.6 (buffer D) followed by another wash with 100 mM H3COONa pH 4.0. Bound protein was eluted with 100 mM H3COONa pH 4.0.
  • the peak fractions were pooled and loaded at 4 ml/min onto a 5 ml HiTrap S column equilibrated with 100 mM H 3 COONa pH 5.5, and washed with the equilibration buffer followed by elution with a gradient from 0-1 M NaCl in 50 mM NaPO 4 pH 5.5. Peak fractions eluting at about 500 mM NaCl were pooled.
  • DC activation is not a general property of anti-DC-ASGPR antibodies, rather only certain anti-DC-ASGPR mAbs have this function.
  • FIG. 5 shows that only certain mAbs activate DCS through the DC-ASGPR, which must be characterized by screening against actual DCs.
  • anti-DC-ASGPR mAbs encompasses particular amino acid sequences shown below corresponding to anti-DC-ASGPR monoclonal antibodies that are desirable components (in the context of e.g., humanized recombinant antibodies) of therapeutic or protective products. The following are such sequences in the context of chimeric mouse V region—human C region recombinant antibodies. [mAnti-ASGPR_49C11_7H-LV-hIgG4H-C] is
  • V-region sequences and related sequences modified by those well versed in the art to e.g., enhance affinity for DC-ASGPR and/or integrated into human V-region framework sequences to be engineered into expression vectors to direct the expression of protein forms that can bind to DC-ASGPR on antigen presenting cells.
  • Engineered recombinant anti-DC-ASGPR recombinant antibody—antigen fusion proteins ((rAb.antigen) are efficacious prototype vaccines in vitro—Expression vectors can be constructed with diverse protein coding sequence e.g., fused in-frame to the H chain coding sequence.
  • antigens such as Influenza HAS, Influenza M1, HIV gag, or immuno-dominant peptides from cancer antigens, or cytokines
  • rAb.antigen or rAb.cytokine fusion proteins which in the context of this invention, can have utility derived from using the anti-DC-ASGPR V-region sequence to bring the antigen or cytokine (or toxin) directly to the surface of the antigen presenting cell bearing DC-AS GPR.
  • An exemplative prototype vaccine based on this concept could use a H chain vector such as [mAnti-ASGPR_5F10H-LV-hIgG4H-C-Flex-FluHA5-1-6 ⁇ His] or
  • FIG. 5 shows that certain anti-DC-ASGPR mAbs can activate DC.
  • DC were incubated for 24 hrs with one of a panel of 12 pure anti-ASGPR mAbs.
  • Cells were then tested for expression of cell surface CD86 (a DC activation marker) and supernatants were assayed for secreted cytokines.
  • Three mAbs (36, 38, 43) from the anti-ASGPR mAb panel activated DC.
  • FIG. 6 shows that different antigens can be expressed in the context of a DC-ASGPR rAb.
  • Such an anti-DC-ASGPR rAb.Doc protein can be simply mixed with any Cohesin.fusion protein to assemble a stable non-covalent [rAb.Doc:Coh.fusion] complex that functions just as a rAb.fusion protein.
  • FIG. 6 shows that such a [rAb.Doc:Coh.fusion] complex can focus antigen to the surface of cells expressing DC-ASGPR.
  • the figure also shows anti-DC-ASGPR.Doc:Coh.Flu M1 complexes deliver Flu M1 to the surface of 293F cells transfected with DC-ASGPR cDNA.
  • Anti-DC-ASGPR rAb complexed to Flu M1 via Dockerin:Cohesin interaction targets the antigen to human DCs and results in the expansion of Flu M1-specific CD8+ T cells—the potential utility of anti-DC-ASGPR rAbs as devices to deliver antigen to e.g., DC is shown in the figure below.
  • FIG. 7 shows the dramatic expansion of Flu M1-specific CD8+ cells is highly predictive of potency of such an agent as a vaccine directed to eliciting protective immune responses against Flu M1.
  • FIGS. 8A-8D demonstrated the cross reactivity of the different antibodies with monkey ASGPR.
  • pIRES_ ASGPR-mon monkey
  • the sequence of final product is base on clone 5S10.
  • Most other clones are either similar to this with one aa difference or identical to this.
  • one clone, 5S1 has an A deletion near the 3′ end, which generated a shortened and different C′ terminus and maybe used as a second variant.
  • DC-ASGPR_MoN gaattcgctagcCACCATGACATATGAAAACTTCCAAGACTTGGAGAGTGAGGAAAGT CCAAGGGG (SEQ ID NO.: 15); and DC-ASGPR_Mo: CGAATTCGCGGCCGCTCAGTGACTCTCCTGGCTGGCCTGGGTCAGACCAGCCTCGC AGACCC (SEQ ID NO.: 16), which is a reverse complement of GGGTCTGCGAGGCTGGTCTGACCCAGGCCAGCCAGGAGAGTCACTGAGCGGCCGC GAATTCG (SEQ ID NO.: 17). Sequence comparisons indicate the likely regions of overlap and, hence, the cross-reactivity, as is known to those if skill in the art.
  • compositions of the invention can be used to achieve methods of the invention.
  • the words “comprising” (and any form of comprising, such as “comprise” and “comprises”), “having” (and any form of having, such as “have” and “has”), “including” (and any form of including, such as “includes” and “include”) or “containing” (and any form of containing, such as “contains” and “contain”) are inclusive or open-ended and do not exclude additional, unrecited elements or method steps.
  • A, B, C, or combinations thereof refers to all permutations and combinations of the listed items preceding the term.
  • “A, B, C, or combinations thereof” is intended to include at least one of: A, B, C, AB, AC, BC, or ABC, and if order is important in a particular context, also BA, CA, CB, CBA, BCA, ACB, BAC, or CAB.
  • expressly included are combinations that contain repeats of one or more item or term, such as BB, AAA, MB, BBC, AAABCCCC, CBBAAA, CABABB, and so forth.
  • BB BB
  • AAA AAA
  • MB BBC
  • AAABCCCCCC CBBAAA
  • CABABB CABABB
  • compositions and/or methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. While the compositions and methods of this invention have been described in terms of preferred embodiments, it will be apparent to those of skill in the art that variations may be applied to the compositions and/or methods and in the steps or in the sequence of steps of the method described herein without departing from the concept, spirit and scope of the invention. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention as defined by the appended claims.

Abstract

The present invention includes compositions and methods for making and using anti DC-ASGPR antibodies that can, e.g., activate DCs and other cells.

Description

CROSS-REFERENCE TO RELATED APPLICATIONS
This application is a continuation of U.S. patent application Ser. No. 15/265,236, filed Sep. 14, 2016, which is a continuation application of U.S. patent application Ser. No. 14/254,206, filed Apr. 16, 2014, now U.S. Pat. No. 9,453,074, issued Sep. 27, 2016, which is a continuation application of U.S. patent application Ser. No. 13/551,198, filed Jul. 17, 2012, now U.S. Pat. No. 8,728,481, issued May 20, 2014, which is a divisional of U.S. patent application Ser. No. 12/025,010 filed Feb. 2, 2008, now U.S. Pat. No. 8,236,934, issued Aug. 7, 2012, which claims priority to U.S. Provisional Application Ser. No. 60/888,036, filed Feb. 2, 2007, the contents of each of which are incorporated by reference herein in their entirety.
STATEMENT OF FEDERALLY FUNDED RESEARCH
This invention was made with government support under Grant No. U19 AI057234 awarded by the National Institutes of Health (NIH). The government has certain rights in the invention.
TECHNICAL FIELD OF THE INVENTION
The present invention relates in general to the field of agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (DC-ASGPR).
REFERENCE TO A SEQUENCE LISTING
The present application includes a Sequence Listing filed separately as required by 37 CFR 1.821-1.825.
BACKGROUND OF THE INVENTION
Without limiting the scope of the invention, its background is described in connection with antigen presentation.
Dendritic Cells play a pivotal role in controlling the interface of innate and acquired immunity by providing soluble and intercellular signals, followed by recognition of pathogens. These functions of DCs are largely dependent on the expression of specialized surface receptors, ‘pattern recognition receptors’ (PRRs), represented, most notably, by toll-like receptors (TLRs) and C-type lectins or lectin-like receptors (LLRs) (1-3).
In the current paradigm, a major role of TLRs is to alert DCs to produce interleukin 12 (IL-12) and other inflammatory cytokines for initiating immune responses. C-type LLRs operate as constituents of the powerful antigen capture and uptake mechanism of macrophages and DCs (1). Compared to TLRs, however, LLRs might have broader ranges of biological functions that include cell migrations (4), intercellular interactions (5). These multiple functions of LLRs might be due to the facts that LLRs, unlike TLRs, can recognize both self and non-self. However, the complexity of LLRs, including the redundancy of a number of LLRs expressed in immune cells, has been one of the major obstacles to understand the detailed functions of individual LLRs. In addition, natural ligands for most of these receptors remain unidentified. Nonetheless, evidence from recent studies suggests that LLRs, in collaboration with TLRs, may contribute to the activation of immune cells during microbial infections (6-14).
Valladeau et al. (The Journal of Immunology, 2001, 167: 5767-5774) described a novel LLR receptor on immature human Dendritic Cells related to hepatic Asialoglycoprotein Receptor and demonstrated that it efficiently mediated endocytosis. DC-ASGPR mRNA was observed predominantly in immune tissues—in DC and granulocytes, but not in T, B, or NK cells, or monocytes. DC-ASGPR species were restricted to the CD14-derived DC obtained from CD34-derived progenitors, while absent from the CD1a-derived subset. Both monocyte-derived DC and tonsillar interstitial-type DC expressed DC-ASGPR protein, while Langerhans-type cells did not. Furthermore, DC-ASGPR was a feature of immaturity, as expression was lost upon CD40 activation. In agreement with the presence of tyrosine-based and dileucine motifs in the intracytoplasmic domain, mAb against DC-ASGPR was rapidly internalized by DC at 37° C. Finally, intracellular DC-ASGPR was localized to early endosomes, suggesting that the receptor recycles to the cell surface following internalization of ligand. These findings identified DC-ASGPR/human macrophage lectin as a feature of immature DC, and as another lectin important for the specialized Ag-capture function of DC.
SUMMARY OF THE INVENTION
While DC-ASGPR is known to be capable of directing the internalization of surrogate antigen into human DC, the invention uses novel biological activities of DC-ASGPR to effect particularly desirable changes in the immune system, some in the context of antigen uptake (e.g., vaccination), others through the unique action of DC-ASGPR effectors (alone or in concert with other immune regulatory molecules) capable of eliciting signaling through this receptor on DC, B cells, and monocytes. The invention disclosure reveals means of developing unique agents capable of activating cells bearing DC-ASGPR, as well as the effect of the resulting changes in cells receiving these signals regards action on other cells in the immune system. These effects (either alone, or in concert with other signals (i.e., co-stimulation)) are highly predictive of therapeutic outcomes for certain disease states or for augmenting protective outcomes in the context of vaccination.
The present invention includes compositions and methods for increasing the effectiveness of antigen presentation by a DC-ASGPR-expressing antigen presenting cell by isolating and purifying a DC-ASGPR-specific antibody or fragment thereof to which a targeted agent is attached that forms an antibody-antigen complex, wherein the agent is processed and presented by, e.g., a dendritic cell, that has been contacted with the antibody-agent complex. In one embodiment, the antigen presenting cell is a dendritic cell and the DC-ASGPR-specific antibody or fragment thereof is bound to one half of a Coherin/Dockerin pair. The DC-ASGPR-specific antibody or fragment thereof may also be bound to one half of a Coherin/Dockerin pair and an antigen is bound to the complementary half of the Coherin/Dockerin pair to form a complex. Non-limiting examples agents include one or more peptides, proteins, lipids, carbohydrates, nucleic acids and combinations thereof.
The agent may one or more cytokine selected from interleukins, transforming growth factors (TGFs), fibroblast growth factors (FGFs), platelet derived growth factors (PDGFs), epidermal growth factors (EGFs), connective tissue activated peptides (CTAPs), osteogenic factors, and biologically active analogs, fragments, and derivatives of such growth factors, B/T-cell differentiation factors, B/T-cell growth factors, mitogenic cytokines, chemotactic cytokines, colony stimulating factors, angiogenesis factors, IFN-α, IFN-β, IFN-γ, IL1, IL2, IL3, IL4, IL5, IL6, IL7, IL8, IL9, IL10, IL11, IL12, IL13, IL14, IL15, IL16, IL17, IL18, etc., leptin, myostatin, macrophage stimulating protein, platelet-derived growth factor, TNF-α, TNF-β, NGF, CD40L, CD137L/4-1BBL, human lymphotoxin-β, G-CSF, M-CSF, GM-CSF, PDGF, IL-1α, IL1-β, IP-10, PF4, GRO, 9E3, erythropoietin, endostatin, angiostatin, VEGF, transforming growth factor (TGF) supergene family include the beta transforming growth factors (for example TGF-β1, TGF-β2, TGF-β3); bone morphogenetic proteins (for example, BMP-1, BMP-2, BMP-3, BMP-4, BMP-5, BMP-6, BMP-7, BMP-8, BMP-9); heparin-binding growth factors (fibroblast growth factor (FGF), epidermal growth factor (EGF), platelet-derived growth factor (PDGF), insulin-like growth factor (IGF)); Inhibins (for example, Inhibin A, Inhibin B); growth differentiating factors (for example, GDF-1); and Activins (for example, Activin A, Activin B, Activin AB). In another embodiment, the agent comprises an antigen that is a bacterial, viral, fungal, protozoan or cancer protein.
The present invention also includes compositions and methods for increasing the effectiveness of antigen presentation by dendritic cells comprising binding a DC-ASGPR-specific antibody or fragment thereof to which an antigen is attached that forms an antibody-antigen complex, wherein the antigen is processed and presented by a dendritic cell that has been contacted with the antibody-antigen complex. Another embodiment is the use of antibodies or other specific binding molecules directed to DC-AS GPR for delivering antigens to antigen-presenting cells for the purpose of eliciting protective or therapeutic immune responses. The use of antigen-targeting reagents specific to DC-ASGPR for vaccination via the skin; antigen-targeting reagents specific to DC-AS GPR in association with co-administered or linked adjuvant for vaccination or use for antigen-targeting (vaccination) purposes of specific antigens which can be expressed as recombinant antigen-antibody fusion proteins.
Another embodiment includes a method for increasing the effectiveness of dendritic cells by isolating patient dendritic cells; exposing the dendritic cells to activating amounts of anti-DC-ASGPR antibodies or fragments thereof and antigen to form antigen-loaded, activated dendritic cells; and reintroducing the antigen-loaded, activated dendritic cells into the patient. The antigen may be a bacterial, viral, fungal, protozoan or cancer protein. The present invention also includes an anti-DC-AS GPR immunoglobulin or portion thereof that is secreted from mammalian cells and an antigen bound to the immunoglobulin. The immunoglobulin is bound to one half of a cohesin/dockerin domain, or it may also include a complementary half of the cohesin-dockerin binding pair bound to an antigen that forms a complex with the modular rAb carrier, or a complementary half of the cohesin-dockerin binding pair that is a fusion protein with an antigen. The antigen specific domain may be a full length antibody, an antibody variable region domain, an Fab fragment, a Fab′ fragment, an F(ab)2 fragment, and Fv fragment, and Fabc fragment and/or a Fab fragment with portions of the Fc domain. The anti-DC-ASGPR immunoglobulin may also be bound to a toxin selected from wherein the toxin is selected from the group consisting of a radioactive isotope, metal, enzyme, botulin, tetanus, ricin, cholera, diphtheria, aflatoxins, perfringens toxin, mycotoxins, shigatoxin, staphylococcal enterotoxin B, T2, seguitoxin, saxitoxin, abrin, cyanoginosin, alphatoxin, tetrodotoxin, aconotoxin, snake venom and spider venom. The antigen may be a fusion protein with the immunoglobulin or bound chemically covalently or not.
The present invention also includes compositions and methods for increasing the effectiveness of dendritic cells by isolating patient dendritic cells, exposing the dendritic cells to activating amounts of anti-DC-ASGPR antibodies or fragments thereof and antigen to form antigen-loaded, activated dendritic cells; and reintroducing the antigen-loaded, activated dendritic cells into the patient. The agents may be used to engage DC-ASGPR, alone or with co-activating agents, to activate antigen-presenting cells for therapeutic or protective applications, to bind DC-ASGPR and/or activating agents linked to antigens, alone or with co-activating agents, for protective or therapeutic vaccination. Another use of is the development of specific antibody V-region sequences capable of binding to and activating DC-ASGPR, for use as anti-DC-ASGPR agents linked to toxic agents for therapeutic purposes in the context of diseases known or suspected to result from inappropriate activation of immune cells via DC-ASGPR and as a vaccine with a DC-ASGPR-specific antibody or fragment thereof to which an antigen is attached that forms an antibody-antigen complex, wherein the antigen is processed and presented by a dendritic cell that has been contacted with the antibody-antigen complex.
BRIEF DESCRIPTION OF THE DRAWINGS
For a more complete understanding of the features and advantages of the present invention, reference is now made to the detailed description of the invention along with the accompanying figures and in which:
FIGS. 1A to 1E demonstrate signaling through lectin-like receptor DC-AS GPR activates DCs, resulting in increased levels of costimulatory molecules as well as cytokines and chemokines. FIG. 1A shows three day and six day GM/IL-4 DCs were stained with FITC-labeled goat anti-mouse IgG followed by mouse monoclonal anti-human DC-ASGPR, antibody.
FIG. 1B shows six day GM/IL-4 DCs were cultured in plates coated with the anti-DC-ASGPR or control mAbs (1-2 ug/ml) for 16-18 h. Cells were stained with anti-CD86 and HLA-DR antibodies labeled with fluorescent dyes. Open and filled bars in the histograms represent cells activated with isotype control mAbs and anti-lectin mAbs, respectively. FIG. 1C shows six day GM/IL-4 DCs were cultured in plates coated with the mAbs for 12 h, and subjected to RNA isolation and Affymetrix Gene Chip analysis, as described in Methods. Fold increases of gene expression by anti-lectin mAbs were compared with the gene expression levels in DCs stimulated with control mAbs. FIG. 1D shows the cytokines and chemokines in the culture supernatants from the experiment shown in FIG. 1B were measured by Luminex. FIG. 1E shows six day GM/IL-4 DCs were cultured in plates coated with mAbs in the presence or absence of 50 ng/ml soluble CD40L, for 16-18 h, and then stained with anti-CD83 antibodies. Cytokines and chemokines in the culture supernatants from the experiment shown in FIG. 1E were measured by Luminex. Results shown are representative of three independent experiments using cells from different normal donors.
FIGS. 2A to 2D shows that DC-ASGPR expressed on DCs, contributes to enhanced humoral immune responses. Six day GM/IL-4 DCs, 5×103/well, were incubated in 96 well plates coated with anti-DC-ASGPR or control mAb for 16-18 h, and then 1×105 autologous CD19+ B cells stained with CFSE were co-cultured in the presence of 20 units/ml IL-2 and 50 nM CpG. FIG. 2A is a FACS of day six cells stained with fluorescently labeled antibodies. CD3+ and 7-AAD+ cells were gated out. CD38+ and CFSE cells were purified by FACS sorter and Giemsa staining was performed. FIG. 2B are culture supernatants on day thirteen were analyzed for total IgM, IgG, and IgM by sandwich ELISA. FIG. 1C shows DCs pulsed with 5 multiplicity of infection (moi) of heat-inactivated influenza virus (PR8), and cultured with B cells. Culture supernatant was analyzed for influenza-specific immunoglobulins (Igs) on day thirteen. FIG. 1D shows DC cultured with anti-DC-ASGPR or control mAb were stained for cell surface APRIL expression and the supernatants assayed for soluble APRIL.
FIGS. 3A to 3D shows the cell surface expression of DC-ASGPR on B cells contribute to B cell activation and immunoglobulin production. FIG. 3A are PBMCs from buffy coats were stained with anti-CD19, anti-CD3, and anti-DC-ASGPR or control mAb. CD19+ and CD3+ cells were gated and the expression levels of the molecules on CD19+ B cells were measured by flow cytometry. FIG. 3B are CD19+ B cells from buffy coats were cultured in plates coated with the mAbs for 12 h, and subjected to RNA isolation and Affymetrix Gene Chip analysis as described in Methods. Fold increases of gene expression by anti-DC-ASGPR mAb were compared to the gene expression levels in CD19+ B cells stimulated with control mAb.
FIG. 3C shows CD19+ B cells were cultured in plates coated with the mAbs for 16-18 h, and then culture supernatants were analyzed for cytokines and chemokines by Luminex. FIG. 3D shows 1×105 CD19+ B cells were cultured in plates coated with the mAbs for thirteen days. Total Ig levels were measured by ELISA. Data are representative of two repeat experiments using cells from three different normal donors.
FIGS. 4A to 4D shows that the proliferation of purified allogeneic T cells was significantly enhanced by DCs stimulated with mAb specific for DC-ASGPR.
FIG. 5 shows that certain anti-DC-ASGPR mAbs can activate DC. GM-CSF/IL-4. DC were incubated for 24 hrs with one of a panel of 12 pure anti-ASGPR mAbs. Cells were then tested for expression of cell surface CD86 (a DC activation marker) and supernatants were assayed for secreted cytokines. Three mAbs (36, 38, 43) from the anti-ASGPR mAb panel activated DC.
FIG. 6 shows that different antigens can be expressed in the context of a DC-ASGPR rAb. Such an anti-DC-ASGPR rAb.Doc protein can be simply mixed with any Cohesin.fusion protein to assemble a stable non-covalent [rAb.Doc:Coh.fusion] complex that functions just as a rAb.fusion protein.
FIG. 7—GM-CSF/IFNa DCs (5,000/well) were loaded with 10 or 1 nM anti-DC-ASGPR.Doc:Coh.Flu M1, or hIgG4.Doc:Coh.Flu M1 complexes. After 6 h, autologous CD8+ T cells (200,000/well) were added into the cultures. At day 8, the CD8+ T cells were analyzed for expansion of cells bearing TCR specific for a HLA-A201 immuno-dominant peptide. The inner boxes indicate the percentage of tetramer-specific CD8+ T cells.
FIGS. 8A-8D demonstrated the cross reactivity of the different antibodies with monkey ASGPR.
DETAILED DESCRIPTION OF THE INVENTION
While the making and using of various embodiments of the present invention are discussed in detail below, it should be appreciated that the present invention provides many applicable inventive concepts that can be embodied in a wide variety of specific contexts. The specific embodiments discussed herein are merely illustrative of specific ways to make and use the invention and do not delimit the scope of the invention.
To facilitate the understanding of this invention, a number of terms are defined below. Terms defined herein have meanings as commonly understood by a person of ordinary skill in the areas relevant to the present invention. Terms such as “a”, “an” and “the” are not intended to refer to only a singular entity, but include the general class of which a specific example may be used for illustration. The terminology herein is used to describe specific embodiments of the invention, but their usage does not delimit the invention, except as outlined in the claims.
Dendritic cells (DCs) are antigen-presenting cells that play a key role in regulating antigen-specific immunity (Mellman and Steinman 2001), (Banchereau, Briere et al. 2000), (Cella, Sallusto et al. 1997). DCs capture antigens, process them into peptides, and present these to T cells. Therefore delivering antigens directly to DC is a focus area for improving vaccines. One such example is the development of DC-based vaccines using ex-vivo antigen-loading of autologous DCs that are then re-administrated to patients (Banchereau, Schuler-Thurner et al. 2001), (Steinman and Dhodapkar 2001). Another strategy to improve vaccine efficacy is specific targeting to DC of antigen conjugated to antibodies against internalizing DC-specific receptors. The potential of targeting DCfor vaccination is highlighted by key mouse studies. In vivo, targeting with an anti-LOX-1 mAb coupled to ovalbumin (OVA) induced a protective CD8+ T cell response, via exogenous antigen cross-presentation toward the MHC class I pathway (Delneste, Magistrelli et al. 2002). Also, OVA conjugated to anti-DEC205 mAb in combination with a CD40L maturation stimulus enhanced the MHC class I-restricted presentation by DCs in vivo and led to the durable formation of effector memory CD8+ T cells (Bonifaz, Bonnyay et al. 2004). Both these studies showed dramatic dose-sparing (i.e., strong immune-responses at very low antigen doses) and suggested broader responses than normally seen with other types of OVA immunization. Recent work with targeting of HIV gag antigen to DC via DEC205 has extended these concepts to a clinically relevant antigen and confirmed the tenents of targeting antigen to DC—dramatic dose-sparing, protective responses from a single vaccination, and expansion of antigen-specific T cells in both the CD8 and CD4 compartments (Trumpfheller, Finke et al. 2006).
The present invention provides for the complexing of multiple antigens or proteins (engineered, expressed, and purified independently from the primary mAb) in a controlled, multivariable fashion, to one single primary recombinant mAb. Presently, there are methods for engineering site-specific biotinylation sites that provide for the addition of different proteins (each engineered separately linked to streptavidin) to the one primary mAb. However, the present invention provides for addition to the primary mAb of multiple combinations, in fixed equimolar ratios and locations, of separately engineered proteins.
As used herein, the term “modular rAb carrier” is used to describe a recombinant antibody system that has been engineered to provide the controlled modular addition of diverse antigens, activating proteins, or other antibodies to a single recombinant monoclonal antibody (mAb). The rAb may be a monoclonal antibody made using standard hybridoma techniques, recombinant antibody display, humanized monoclonal antibodies and the like. The modular rAb carrier can be used to, e.g., target (via one primary recombinant antibody against an internalizing receptor, e.g., a human dendritic cell receptor) multiple antigens and/or antigens and an activating cytokine to dendritic cells (DC). The modular rAb carrier may also be used to join two different recombinant mAbs end-to-end in a controlled and defined manner.
The antigen binding portion of the “modular rAb carrier” may be one or more variable domains, one or more variable and the first constant domain, an Fab fragment, a Fab′ fragment, an F(ab)2 fragment, and Fv fragment, and Fabc fragment and/or a Fab fragment with portions of the Fc domain to which the cognate modular binding portions are added to the amino acid sequence and/or bound. The antibody for use in the modular rAb carrier can be of any isotype or class, subclass or from any source (animal and/or recombinant).
In one non-limiting example, the modular rAb carrier is engineered to have one or more modular cohesin-dockerin protein domains for making specific and defined protein complexes in the context of engineered recombinant mAbs. The mAb is a portion of a fusion protein that includes one or more modular cohesin-dockerin protein domains carboxy from the antigen binding domains of the mAb. The cohesin-dockerin protein domains may even be attached post-translationally, e.g., by using chemical cross-linkers and/or disulfide bonding.
The term “antigen” as used herein refers to a molecule that can initiate a humoral and/or cellular immune response in a recipient of the antigen. Antigen may be used in two different contexts with the present invention: as a target for the antibody or other antigen recognition domain of the rAb or as the molecule that is carried to and/or into a cell or target by the rAb as part of a dockerin/cohesin-molecule complement to the modular rAb carrier. The antigen is usually an agent that causes a disease for which a vaccination would be advantageous treatment. When the antigen is presented on MHC, the peptide is often about 8 to about 25 amino acids. Antigens include any type of biologic molecule, including, for example, simple intermediary metabolites, sugars, lipids and hormones as well as macromolecules such as complex carbohydrates, phospholipids, nucleic acids and proteins. Common categories of antigens include, but are not limited to, viral antigens, bacterial antigens, fungal antigens, protozoal and other parasitic antigens, tumor antigens, antigens involved in autoimmune disease, allergy and graft rejection, and other miscellaneous antigens.
The modular rAb carrier is able to carry any number of active agents, e.g., antibiotics, anti-infective agents, antiviral agents, anti-tumoral agents, antipyretics, analgesics, anti-inflammatory agents, therapeutic agents for osteoporosis, enzymes, cytokines, anticoagulants, polysaccharides, collagen, cells, and combinations of two or more of the foregoing active agents. Examples of antibiotics for delivery using the present invention include, without limitation, tetracycline, aminoglycosides, penicillins, cephalosporins, sulfonamide drugs, chloramphenicol sodium succinate, erythromycin, vancomycin, lincomycin, clindamycin, nystatin, amphotericin B, amantidine, idoxuridine, p-amino salicyclic acid, isoniazid, rifampin, antinomycin D, mithramycin, daunomycin, adriamycin, bleomycin, vinblastine, vincristine, procarbazine, imidazole carboxamide, and the like.
Examples of anti-tumor agents for delivery using the present invention include, without limitation, doxorubicin, Daunorubicin, taxol, methotrexate, and the like. Examples of antipyretics and analgesics include aspirin, Motrin®, Ibuprofen®, naprosyn, acetaminophen, and the like.
Examples of anti-inflammatory agents for delivery using the present invention include, without limitation, include NSAIDS, aspirin, steroids, dexamethasone, hydrocortisone, prednisolone, Diclofenac Na, and the like.
Examples of therapeutic agents for treating osteoporosis and other factors acting on bone and skeleton include for delivery using the present invention include, without limitation, calcium, alendronate, bone GLa peptide, parathyroid hormone and its active fragments, histone H4-related bone formation and proliferation peptide and mutations, derivatives and analogs thereof.
Examples of enzymes and enzyme cofactors for delivery using the present invention include, without limitation, pancrease, L-asparaginase, hyaluronidase, chymotrypsin, trypsin, tPA, streptokinase, urokinase, pancreatin, collagenase, trypsinogen, chymotrypsinogen, plasminogen, streptokinase, adenyl cyclase, superoxide dismutase (SOD), and the like.
Examples of cytokines for delivery using the present invention include, without limitation, interleukins, transforming growth factors (TGFs), fibroblast growth factors (FGFs), platelet derived growth factors (PDGFs), epidermal growth factors (EGFs), connective tissue activated peptides (CTAPs), osteogenic factors, and biologically active analogs, fragments, and derivatives of such growth factors. Cytokines may be B/T-cell differentiation factors, B/T-cell growth factors, mitogenic cytokines, chemotactic cytokines, colony stimulating factors, angiogenesis factors, IFN-α, IFN-β, IFN-γ, IL1, IL2, IL3, IL4, IL5, IL6, IL7, IL8, IL9, IL10, IL11, IL12, IL13, IL14, IL15, IL16, IL17, IL18, etc., leptin, myostatin, macrophage stimulating protein, platelet-derived growth factor, TNF-α, TNF-β, NGF, CD40L, CD137L/4-1BBL, human lymphotoxin-β, G-CSF, M-CSF, GM-CSF, PDGF, IL-1α, IL1-β, IP-10, PF4, GRO, 9E3, erythropoietin, endostatin, angiostatin, VEGF or any fragments or combinations thereof. Other cytokines include members of the transforming growth factor (TGF) supergene family include the beta transforming growth factors (for example TGF-β1, TGF-β2, TGF-β3); bone morphogenetic proteins (for example, BMP-1, BMP-2, BMP-3, BMP-4, BMP-5, BMP-6, BMP-7, BMP-8, BMP-9); heparin-binding growth factors (for example, fibroblast growth factor (FGF), epidermal growth factor (EGF), platelet-derived growth factor (PDGF), insulin-like growth factor (IGF)); Inhibins (for example, Inhibin A, Inhibin B); growth differentiating factors (for example, GDF-1); and Activins (for example, Activin A, Activin B, Activin AB).
Examples of growth factors for delivery using the present invention include, without limitation, growth factors that can be isolated from native or natural sources, such as from mammalian cells, or can be prepared synthetically, such as by recombinant DNA techniques or by various chemical processes. In addition, analogs, fragments, or derivatives of these factors can be used, provided that they exhibit at least some of the biological activity of the native molecule. For example, analogs can be prepared by expression of genes altered by site-specific mutagenesis or other genetic engineering techniques.
Examples of anticoagulants for delivery using the present invention include, without limitation, include warfarin, heparin, Hirudin, and the like. Examples of factors acting on the immune system include for delivery using the present invention include, without limitation, factors which control inflammation and malignant neoplasms and factors which attack infective microorganisms, such as chemotactic peptides and bradykinins.
Examples of viral antigens include, but are not limited to, e.g., retroviral antigens such as retroviral antigens from the human immunodeficiency virus (HIV) antigens such as gene products of the gag, pol, and env genes, the Nef protein, reverse transcriptase, and other HIV components; hepatitis viral antigens such as the S, M, and L proteins of hepatitis B virus, the pre-S antigen of hepatitis B virus, and other hepatitis, e.g., hepatitis A, B, and C, viral components such as hepatitis C viral RNA; influenza viral antigens such as hemagglutinin and neuraminidase and other influenza viral components; measles viral antigens such as the measles virus fusion protein and other measles virus components; rubella viral antigens such as proteins E1 and E2 and other rubella virus components; rotaviral antigens such as VP7sc and other rotaviral components; cytomegaloviral antigens such as envelope glycoprotein B and other cytomegaloviral antigen components; respiratory syncytial viral antigens such as the RSV fusion protein, the M2 protein and other respiratory syncytial viral antigen components; herpes simplex viral antigens such as immediate early proteins, glycoprotein D, and other herpes simplex viral antigen components; varicella zoster viral antigens such as gpl, gpII, and other varicella zoster viral antigen components; Japanese encephalitis viral antigens such as proteins E, M-E, M-E-NS1, NS1, NS1-NS2A, 80% E, and other Japanese encephalitis viral antigen components; rabies viral antigens such as rabies glycoprotein, rabies nucleoprotein and other rabies viral antigen components. See Fundamental Virology, Second Edition, eds. Fields, B. N. and Knipe, D. M. (Raven Press, New York, 1991) for additional examples of viral antigens.
Antigenic targets that may be delivered using the rAb-DC/DC-antigen vaccines of the present invention include genes encoding antigens such as viral antigens, bacterial antigens, fungal antigens or parasitic antigens. Viruses include picornavirus, coronavirus, togavirus, flavirvirus, rhabdovirus, paramyxovirus, orthomyxovirus, bunyavirus, arenavirus, reovirus, retrovirus, papilomavirus, parvovirus, herpesvirus, poxvirus, hepadnavirus, and spongiform virus. Other viral targets include influenza, herpes simplex virus 1 and 2, measles, dengue, smallpox, polio or HIV. Pathogens include trypanosomes, tapeworms, roundworms, helminthes, and malaria. Tumor markers, such as fetal antigen or prostate specific antigen, may be targeted in this manner. Other examples include: HIV env proteins and hepatitis B surface antigen. Administration of a vector according to the present invention for vaccination purposes would require that the vector-associated antigens be sufficiently non-immunogenic to enable long term expression of the transgene, for which a strong immune response would be desired. In some cases, vaccination of an individual may only be required infrequently, such as yearly or biennially, and provide long term immunologic protection against the infectious agent. Specific examples of organisms, allergens and nucleic and amino sequences for use in vectors and ultimately as antigens with the present invention may be found in U.S. Pat. No. 6,541,011, relevant portions incorporated herein by reference, in particular, the tables that match organisms and specific sequences that may be used with the present invention.
Bacterial antigens for use with the rAb vaccine disclosed herein include, but are not limited to, e.g., bacterial antigens such as pertussis toxin, filamentous hemagglutinin, pertactin, FIM2, FIM3, adenylate cyclase and other pertussis bacterial antigen components; diptheria bacterial antigens such as diptheria toxin or toxoid and other diptheria bacterial antigen components; tetanus bacterial antigens such as tetanus toxin or toxoid and other tetanus bacterial antigen components; streptococcal bacterial antigens such as M proteins and other streptococcal bacterial antigen components; gram-negative bacilli bacterial antigens such as lipopolysaccharides and other gram-negative bacterial antigen components, Mycobacterium tuberculosis bacterial antigens such as mycolic acid, heat shock protein 65 (HSP65), the 30 kDa major secreted protein, antigen 85A and other mycobacterial antigen components; Helicobacter pylori bacterial antigen components; pneumococcal bacterial antigens such as pneumolysin, pneumococcal capsular polysaccharides and other pneumococcal bacterial antigen components; haemophilus influenza bacterial antigens such as capsular polysaccharides and other haemophilus influenza bacterial antigen components; anthrax bacterial antigens such as anthrax protective antigen and other anthrax bacterial antigen components; rickettsiae bacterial antigens such as rompA and other rickettsiae bacterial antigen component. Also included with the bacterial antigens described herein are any other bacterial, mycobacterial, mycoplasmal, rickettsial, or chlamydial antigens. Partial or whole pathogens may also be: haemophilus influenza; Plasmodium falciparum; Neisseria meningitidis; Streptococcus pneumoniae; Neisseria gonorrhoeae; salmonella serotype typhi; shigella; Vibrio cholerae; Dengue Fever; Encephalitides; Japanese Encephalitis; Lyme disease; Yersinia pestis; west nile virus; yellow fever; tularemia; hepatitis (viral; bacterial); RSV (respiratory syncytial virus); HPIV 1 and HPIV 3; adenovirus; small pox; allergies and cancers.
Fungal antigens for use with compositions and methods of the invention include, but are not limited to, e.g., candida fungal antigen components; histoplasma fungal antigens such as heat shock protein 60 (HSP60) and other histoplasma fungal antigen components; cryptococcal fungal antigens such as capsular polysaccharides and other cryptococcal fungal antigen components; coccidiodes fungal antigens such as spherule antigens and other coccidiodes fungal antigen components; and tinea fungal antigens such as trichophytin and other coccidiodes fungal antigen components.
Examples of protozoal and other parasitic antigens include, but are not limited to, e.g., Plasmodium falciparum antigens such as merozoite surface antigens, sporozoite surface antigens, circumsporozoite antigens, gametocyte/gamete surface antigens, blood-stage antigen pf 155/RESA and other plasmodial antigen components; toxoplasma antigens such as SAG-1, p30 and other toxoplasmal antigen components; schistosomae antigens such as glutathione-S-transferase, paramyosin, and other schistosomal antigen components; Leishmania major and other leishmaniae antigens such as gp63, lipophosphoglycan and its associated protein and other leishmanial antigen components; and Trypanosoma cruzi antigens such as the 75-77 kDa antigen, the 56 kDa antigen and other trypanosomal antigen components.
Antigen that can be targeted using the rAb of the present invention will generally be selected based on a number of factors, including: likelihood of internalization, level of immune cell specificity, type of immune cell targeted, level of immune cell maturity and/or activation and the like. Examples of cell surface markers for dendritic cells include, but are not limited to, MHC class I, MHC Class II, B7-2, CD18, CD29, CD31, CD43, CD44, CD45, CD54, CD58, CD83, CD86, CMRF-44, CMRF-56, DCIR and/or ASPGR and the like; while in some cases also having the absence of CD2, CD3, CD4, CD8, CD14, CD15, CD16, CD 19, CD20, CD56, and/or CD57. Examples of cell surface markers for antigen presenting cells include, but are not limited to, MHC class I, MHC Class II, CD40, CD45, B7-1, B7-2, IFN-γ receptor and IL-2 receptor, ICAM-1 and/or Fcγ receptor. Examples of cell surface markers for T cells include, but are not limited to, CD3, CD4, CD8, CD 14, CD20, CD11b, CD16, CD45 and HLA-DR.
Target antigens on cell surfaces for delivery includes those characteristic of tumor antigens typically will be derived from the cell surface, cytoplasm, nucleus, organelles and the like of cells of tumor tissue. Examples of tumor targets for the antibody portion of the present invention include, without limitation, hematological cancers such as leukemias and lymphomas, neurological tumors such as astrocytomas or glioblastomas, melanoma, breast cancer, lung cancer, head and neck cancer, gastrointestinal tumors such as gastric or colon cancer, liver cancer, pancreatic cancer, genitourinary tumors such cervix, uterus, ovarian cancer, vaginal cancer, testicular cancer, prostate cancer or penile cancer, bone tumors, vascular tumors, or cancers of the lip, nasopharynx, pharynx and oral cavity, esophagus, rectum, gall bladder, biliary tree, larynx, lung and bronchus, bladder, kidney, brain and other parts of the nervous system, thyroid, Hodgkin's disease, non-Hodgkin's lymphoma, multiple myeloma and leukemia.
Examples of antigens that may be delivered alone or in combination to immune cells for antigen presentation using the present invention include tumor proteins, e.g., mutated oncogenes; viral proteins associated with tumors; and tumor mucins and glycolipids. The antigens may be viral proteins associated with tumors would be those from the classes of viruses noted above. Certain antigens may be characteristic of tumors (one subset being proteins not usually expressed by a tumor precursor cell), or may be a protein which is normally expressed in a tumor precursor cell, but having a mutation characteristic of a tumor. Other antigens include mutant variant(s) of the normal protein having an altered activity or subcellular distribution, e.g., mutations of genes giving rise to tumor antigens.
Specific non-limiting examples of tumor antigens include: CEA, prostate specific antigen (PSA), HER-2/neu, BAGE, GAGE, MAGE 1-4, 6 and 12, MUC (Mucin) (e.g., MUC-1, MUC-2, etc.), GM2 and GD2 gangliosides, ras, myc, tyrosinase, MART (melanoma antigen), Pmel 17(gp100), GnT-V intron V sequence (N-acetylglucoaminyltransferase V intron V sequence), Prostate Ca psm, PRAME (melanoma antigen), β-catenin, MUM-1-B (melanoma ubiquitous mutated gene product), GAGE (melanoma antigen) 1, BAGE (melanoma antigen) 2-10, c-ERB2 (Her2/neu), EBNA (Epstein-Barr Virus nuclear antigen) 1-6, gp75, human papilloma virus (HPV) E6 and E7, p53, lung resistance protein (LRP), Bcl-2, and Ki-67. In addition, the immunogenic molecule can be an autoantigen involved in the initiation and/or propagation of an autoimmune disease, the pathology of which is largely due to the activity of antibodies specific for a molecule expressed by the relevant target organ, tissue, or cells, e.g., SLE or MG. In such diseases, it can be desirable to direct an ongoing antibody-mediated (i.e., a Th2-type) immune response to the relevant autoantigen towards a cellular (i.e., a Th1-type) immune response. Alternatively, it can be desirable to prevent onset of or decrease the level of a Th2 response to the autoantigen in a subject not having, but who is suspected of being susceptible to, the relevant autoimmune disease by prophylactically inducing a Th1 response to the appropriate autoantigen. Autoantigens of interest include, without limitation: (a) with respect to SLE, the Smith protein, RNP ribonucleoprotein, and the SS-A and SS-B proteins; and (b) with respect to MG, the acetylcholine receptor. Examples of other miscellaneous antigens involved in one or more types of autoimmune response include, e.g., endogenous hormones such as luteinizing hormone, follicular stimulating hormone, testosterone, growth hormone, prolactin, and other hormones.
Antigens involved in autoimmune diseases, allergy, and graft rejection can be used in the compositions and methods of the invention. For example, an antigen involved in any one or more of the following autoimmune diseases or disorders can be used in the present invention: diabetes, diabetes mellitus, arthritis (including rheumatoid arthritis, juvenile rheumatoid arthritis, osteoarthritis, psoriatic arthritis), multiple sclerosis, myasthenia gravis, systemic lupus erythematosis, autoimmune thyroiditis, dermatitis (including atopic dermatitis and eczematous dermatitis), psoriasis, Sjogren's Syndrome, including keratoconjunctivitis sicca secondary to Sjogren's Syndrome, alopecia areata, allergic responses due to arthropod bite reactions, Crohn's disease, aphthous ulcer, iritis, conjunctivitis, keratoconjunctivitis, ulcerative colitis, asthma, allergic asthma, cutaneous lupus erythematosus, scleroderma, vaginitis, proctitis, drug eruptions, leprosy reversal reactions, erythema nodosum leprosum, autoimmune uveitis, allergic encephalomyelitis, acute necrotizing hemorrhagic encephalopathy, idiopathic bilateral progressive sensorineural hearing loss, aplastic anemia, pure red cell anemia, idiopathic thrombocytopenia, polychondritis, Wegener's granulomatosis, chronic active hepatitis, Stevens-Johnson syndrome, idiopathic sprue, lichen planus, Crohn's disease, Graves ophthalmopathy, sarcoidosis, primary biliary cirrhosis, uveitis posterior, and interstitial lung fibrosis. Examples of antigens involved in autoimmune disease include glutamic acid decarboxylase 65 (GAD 65), native DNA, myelin basic protein, myelin proteolipid protein, acetylcholine receptor components, thyroglobulin, and the thyroid stimulating hormone (TSH) receptor. Examples of antigens involved in allergy include pollen antigens such as Japanese cedar pollen antigens, ragweed pollen antigens, rye grass pollen antigens, animal derived antigens such as dust mite antigens and feline antigens, histocompatiblity antigens, and penicillin and other therapeutic drugs. Examples of antigens involved in graft rejection include antigenic components of the graft to be transplanted into the graft recipient such as heart, lung, liver, pancreas, kidney, and neural graft components. The antigen may be an altered peptide ligand useful in treating an autoimmune disease.
As used herein, the term “epitope(s)” refer to a peptide or protein antigen that includes a primary, secondary or tertiary structure similar to an epitope located within any of a number of pathogen polypeptides encoded by the pathogen DNA or RNA. The level of similarity will generally be to such a degree that monoclonal or polyclonal antibodies directed against such polypeptides will also bind to, react with, or otherwise recognize, the peptide or protein antigen. Various immunoassay methods may be employed in conjunction with such antibodies, such as, for example, Western blotting, ELISA, RIA, and the like, all of which are known to those of skill in the art. The identification of pathogen epitopes, and/or their functional equivalents, suitable for use in vaccines is part of the present invention. Once isolated and identified, one may readily obtain functional equivalents. For example, one may employ the methods of Hopp, as taught in U.S. Pat. No. 4,554,101, incorporated herein by reference, which teaches the identification and preparation of epitopes from amino acid sequences on the basis of hydrophilicity. The methods described in several other papers, and software programs based thereon, can also be used to identify epitopic core sequences (see, for example, Jameson and Wolf, 1988; Wolf et al., 1988; U.S. Pat. No. 4,554,101). The amino acid sequence of these “epitopic core sequences” may then be readily incorporated into peptides, either through the application of peptide synthesis or recombinant technology.
The preparation of vaccine compositions that includes the nucleic acids that encode antigens of the invention as the active ingredient, may be prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid prior to infection can also be prepared. The preparation may be emulsified, encapsulated in liposomes. The active immunogenic ingredients are often mixed with carriers which are pharmaceutically acceptable and compatible with the active ingredient.
The term “pharmaceutically acceptable carrier” refers to a carrier that does not cause an allergic reaction or other untoward effect in subjects to whom it is administered. Suitable pharmaceutically acceptable carriers include, for example, one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol, or the like and combinations thereof. In addition, if desired, the vaccine can contain minor amounts of auxiliary substances such as wetting or emulsifying agents, pH buffering agents, and/or adjuvants which enhance the effectiveness of the vaccine. Examples of adjuvants that may be effective include but are not limited to: aluminum hydroxide, N-acetyl-muramyl-L-threonyl-D-isoglutamine (thr-MDP), N-acetyl-nor-muramyl-L-alanyl-D-isoglutamine, MTP-PE and RIB I, which contains three components extracted from bacteria, monophosporyl lipid A, trehalose dimycolate and cell wall skeleton (MPL+TDM+CWS) in a 2% squalene/Tween 80 emulsion. Other examples of adjuvants include DDA (dimethyldioctadecylammonium bromide), Freund's complete and incomplete adjuvants and QuilA. In addition, immune modulating substances such as lymphokines (e.g., IFN-γ, IL-2 and IL-12) or synthetic IFN-γ inducers such as poly I:C can be used in combination with adjuvants described herein.
Pharmaceutical products that may include a naked polynucleotide with a single or multiple copies of the specific nucleotide sequences that bind to specific DNA-binding sites of the apolipoproteins present on plasma lipoproteins as described in the current invention. The polynucleotide may encode a biologically active peptide, antisense RNA, or ribozyme and will be provided in a physiologically acceptable administrable form. Another pharmaceutical product that may spring from the current invention may include a highly purified plasma lipoprotein fraction, isolated according to the methodology, described herein from either the patients blood or other source, and a polynucleotide containing single or multiple copies of the specific nucleotide sequences that bind to specific DNA-binding sites of the apolipoproteins present on plasma lipoproteins, prebound to the purified lipoprotein fraction in a physiologically acceptable, administrable form.
Yet another pharmaceutical product may include a highly purified plasma lipoprotein fraction which contains recombinant apolipoprotein fragments containing single or multiple copies of specific DNA-binding motifs, prebound to a polynucleotide containing single or multiple copies of the specific nucleotide sequences, in a physiologically acceptable administrable form. Yet another pharmaceutical product may include a highly purified plasma lipoprotein fraction which contains recombinant apolipoprotein fragments containing single or multiple copies of specific DNA-binding motifs, prebound to a polynucleotide containing single or multiple copies of the specific nucleotide sequences, in a physiologically acceptable administrable form.
The dosage to be administered depends to a great extent on the body weight and physical condition of the subject being treated as well as the route of administration and frequency of treatment. A pharmaceutical composition that includes the naked polynucleotide prebound to a highly purified lipoprotein fraction may be administered in amounts ranging from 1 μg to 1 mg polynucleotide and 1 μg to 100 mg protein.
Administration of an rAb and rAb complexes a patient will follow general protocols for the administration of chemotherapeutics, taking into account the toxicity, if any, of the vector. It is anticipated that the treatment cycles would be repeated as necessary. It also is contemplated that various standard therapies, as well as surgical intervention, may be applied in combination with the described gene therapy.
Where clinical application of a gene therapy is contemplated, it will be necessary to prepare the complex as a pharmaceutical composition appropriate for the intended application. Generally this will entail preparing a pharmaceutical composition that is essentially free of pyrogens, as well as any other impurities that could be harmful to humans or animals. One also will generally desire to employ appropriate salts and buffers to render the complex stable and allow for complex uptake by target cells.
Aqueous compositions of the present invention may include an effective amount of the compound, dissolved or dispersed in a pharmaceutically acceptable carrier or aqueous medium. Such compositions can also be referred to as inocula. The use of such media and agents for pharmaceutical active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients also can be incorporated into the compositions. The compositions of the present invention may include classic pharmaceutical preparations. Dispersions also can be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
Disease States. Depending on the particular disease to be treated, administration of therapeutic compositions according to the present invention will be via any common route so long as the target tissue is available via that route in order to maximize the delivery of antigen to a site for maximum (or in some cases minimum) immune response. Administration will generally be by orthotopic, intradermal, subcutaneous, intramuscular, intraperitoneal or intravenous injection. Other areas for delivery include: oral, nasal, buccal, rectal, vaginal or topical. Topical administration would be particularly advantageous for treatment of skin cancers. Such compositions would normally be administered as pharmaceutically acceptable compositions that include physiologically acceptable carriers, buffers or other excipients.
Vaccine or treatment compositions of the invention may be administered parenterally, by injection, for example, either subcutaneously or intramuscularly. Additional formulations which are suitable for other modes of administration include suppositories, and in some cases, oral formulations or formulations suitable for distribution as aerosols. In the case of the oral formulations, the manipulation of T-cell subsets employing adjuvants, antigen packaging, or the addition of individual cytokines to various formulation that result in improved oral vaccines with optimized immune responses. For suppositories, traditional binders and carriers may include, for example, polyalkylene glycols or triglycerides; such suppositories may be formed from mixtures containing the active ingredient in the range of 0.5% to 10%, preferably 1%-2%. Oral formulations include such normally employed excipients as, for example, pharmaceutical grades of mannitol, lactose, starch magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, and the like. These compositions take the form of solutions, suspensions, tablets, pills, capsules, sustained release formulations or powders and contain 10%-95% of active ingredient, preferably 25-70%.
The antigen encoding nucleic acids of the invention may be formulated into the vaccine or treatment compositions as neutral or salt forms. Pharmaceutically acceptable salts include the acid addition salts (formed with free amino groups of the peptide) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or with organic acids such as acetic, oxalic, tartaric, maleic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine, and the like.
Vaccine or treatment compositions are administered in a manner compatible with the dosage formulation, and in such amount as will be prophylactically and/or therapeutically effective. The quantity to be administered depends on the subject to be treated, including, e.g., capacity of the subject's immune system to synthesize antibodies, and the degree of protection or treatment desired. Suitable dosage ranges are of the order of several hundred micrograms active ingredient per vaccination with a range from about 0.1 mg to 1000 mg, such as in the range from about 1 mg to 300 mg, and preferably in the range from about 10 mg to 50 mg. Suitable regiments for initial administration and booster shots are also variable but are typified by an initial administration followed by subsequent inoculations or other administrations. Precise amounts of active ingredient required to be administered depend on the judgment of the practitioner and may be peculiar to each subject. It will be apparent to those of skill in the art that the therapeutically effective amount of nucleic acid molecule or fusion polypeptides of this invention will depend, inter alia, upon the administration schedule, the unit dose of antigen administered, whether the nucleic acid molecule or fusion polypeptide is administered in combination with other therapeutic agents, the immune status and health of the recipient, and the therapeutic activity of the particular nucleic acid molecule or fusion polypeptide.
The compositions can be given in a single dose schedule or in a multiple dose schedule. A multiple dose schedule is one in which a primary course of vaccination may include, e.g., 1-10 separate doses, followed by other doses given at subsequent time intervals required to maintain and or reinforce the immune response, for example, at 1-4 months for a second dose, and if needed, a subsequent dose(s) after several months. Periodic boosters at intervals of 1-5 years, usually 3 years, are desirable to maintain the desired levels of protective immunity. The course of the immunization can be followed by in vitro proliferation assays of peripheral blood lymphocytes (PBLs) co-cultured with ESAT6 or ST-CF, and by measuring the levels of IFN-γ released from the primed lymphocytes. The assays may be performed using conventional labels, such as radionucleotides, enzymes, fluorescent labels and the like. These techniques are known to one skilled in the art and can be found in U.S. Pat. Nos. 3,791,932, 4,174,384 and 3,949,064, relevant portions incorporated by reference.
The modular rAb carrier and/or conjugated rAb carrier-(cohesion/dockerin and/or dockerin-cohesin)-antigen complex (rAb-DC/DC-antigen vaccine) may be provided in one or more “unit doses” depending on whether the nucleic acid vectors are used, the final purified proteins, or the final vaccine form is used. Unit dose is defined as containing a predetermined-quantity of the therapeutic composition calculated to produce the desired responses in association with its administration, i.e., the appropriate route and treatment regimen. The quantity to be administered, and the particular route and formulation, are within the skill of those in the clinical arts. The subject to be treated may also be evaluated, in particular, the state of the subject's immune system and the protection desired. A unit dose need not be administered as a single injection but may include continuous infusion over a set period of time. Unit dose of the present invention may conveniently may be described in terms of DNA/kg (or protein/Kg) body weight, with ranges between about 0.05, 0.10, 0.15, 0.20, 0.25, 0.5, 1, 10, 50, 100, 1,000 or more mg/DNA or protein/kg body weight are administered. Likewise the amount of rAb-DC/DC-antigen vaccine delivered can vary from about 0.2 to about 8.0 mg/kg body weight. Thus, in particular embodiments, 0.4 mg, 0.5 mg, 0.8 mg, 1.0 mg, 1.5 mg, 2.0 mg, 2.5 mg, 3.0 mg, 4.0 mg, 5.0 mg, 5.5 mg, 6.0 mg, 6.5 mg, 7.0 mg and 7.5 mg of the vaccine may be delivered to an individual in vivo. The dosage of rAb-DC/DC-antigen vaccine to be administered depends to a great extent on the weight and physical condition of the subject being treated as well as the route of administration and the frequency of treatment. A pharmaceutical composition that includes a naked polynucleotide prebound to a liposomal or viral delivery vector may be administered in amounts ranging from 1 μg to 1 mg polynucleotide to 1 μg to 100 mg protein. Thus, particular compositions may include between about 1 μg, 5 μg, 10 μg, 20 μg, 30 μg, 40 μg, 50 μg, 60 μg, 70 μg, 80 μg, 100 μg, 150 μg, 200 μg, 250 μg, 500 μg, 600 μg, 700 μg, 800 μg, 900 μg or 1,000 μg polynucleotide or protein that is bound independently to 1 μg, 5 μg, 10 μg, 20 μg, 3.0 μg, 40 μg 50 μg, 60 μg, 70 μg, 80 μg, 100 μg, 150 μg, 200 μg, 250 μg, 500 μg, 600 μg, 700 μg, 800 μg, 900 μg, 1 mg, 1.5 mg, 5 mg, 10 mg, 20 mg, 30 mg, 40 mg, 50 mg, 60 mg, 70 mg, 80 mg, 90 mg or 100 mg vector.
The present invention was tested in an in vitro cellular system that measures immune stimulation of human Flu-specific T cells by dendritic cells to which Flu antigen has been targeted. The results shown herein demonstrate the specific expansion of such antigen specific cells at doses of the antigen which are by themselves ineffective in this system.
The present invention may also be used to make a modular rAb carrier that is, e.g., a recombinant humanized mAb (directed to a specific human dendritic cell receptor) complexed with protective antigens from Ricin, Anthrax toxin, and Staphylococcus B enterotoxin. The potential market for this entity is vaccination of all military personnel and stored vaccine held in reserve to administer to large population centers in response to any biothreat related to these agents. The invention has broad application to the design of vaccines in general, both for human and animal use. Industries of interest include the pharmaceutical and biotechnology industries.
The present invention includes compositions and methods, including vaccines, that specifically target (deliver) antigens to antigen-presenting cells (APCs) for the purpose of eliciting potent and broad immune responses directed against the antigen. These compositions evoke protective or therapeutic immune responses against the agent (pathogen or cancer) from which the antigen was derived. In addition the invention creates agents that are directly, or in concert with other agents, therapeutic through their specific engagement of a receptor called DC-ASGPR that is expressed on antigen-presenting cells.
The novel recombinant humanized mAb (directed to the specific human dendritic cell receptor DC-ASGPR) fused through the antibody (Ab) heavy chain to antigens known or suspected to encode protective antigens. These include as examples for vaccination against various agents—hemagglutinins from Influenza H5N1; HIV gag from attenuated toxins from Ricin, Anthrax toxin, and Staphylococcus B enterotoxin; ‘strings’ of antigenic peptides from melanona antigens, etc. The present invention may be used as a preventative or therapeutic vaccination for at risk or infected patients. The invention has broad application for vaccination against many diseases and cancers, both for human and animal use. Industries that can use the present invention include the pharmaceutical and biotechnological.
The present invention can be used to target antigens to APC for vaccination purposes. It is not known which antigen internalizing receptor will be best suited for this purpose. The invention describes particularly advantageous features of DC-ASGPR as for this purpose. Furthermore, the invention shows that engaging DC-ASGPR can be beneficial in the sense of activating the immune system with highly predicted significant therapeutic benefit.
The present invention includes the development of high affinity monoclonal antibodies against human DC-ASGPR. Receptor ectodomain.hIgG (human IgG1Fc) and AP (human placental alkaline phosphatase) fusion proteins were produced for immunization of mice and screening of mAbs, respectively. An expression construct for hDCIR ectodomain.IgG was described previously (Bates, Fournier et al. 1999) and used the mouse SLAM (mSLAM) signal peptide to direct secretion (Bendtsen, Nielsen et al. 2004). An expression vector for hDCIR ectodomain.AP was generated using PCR to amplify AP resides 133-1581 (gbIBC0096471) while adding a proximal in-frame Xho I site and a distal TGA stop codon and Not I site. This Xho I—Not I fragment replaced the IgG coding sequence in the above hDCIR ectodomain.IgG vector. DC-ASGPR ectodomain constructs in the same Ig and AP vector series contained inserts encoding (bp 484-1251, gi153832017). DC-ASGPR fusion proteins were produced using the FreeStyle™ 293 Expression System (Invitrogen) according to the manufacturer's protocol (1 mg total plasmid DNA with 1.3 ml 293 Fectin reagent/L of transfection). For rAb production, equal amounts of vector encoding the H and L chain were co-transfected. Transfected cells are cultured for 3 days, the culture supernatant was harvested and fresh media added with continued incubation for two days. The pooled supernatants were clarified by filtration. Receptor ectodomain.hIgG was purified by HiTrap protein A affinity chromatography with elution by 0.1 M glycine pH 2.7 and then dialyzed versus PBS. rAbs (recombinant antibodies described later) were purified similarly, by using HiTrap MabSelect™ columns. Mouse mAbs were generated by conventional cell fusion technology. Briefly, 6-week-old BALB/c mice were immunized intraperitonealy with 20 μg of receptor ectodomain.hIgGFc fusion protein with Ribi adjuvant, then boosts with 20 μg antigen 10 days and 15 days later. After 3 months, the mice were boosted again three days prior to taking the spleens. Alternately, mice were injected in the footpad with 1-10 μg antigen in Ribi adjuvant every 3-4 days over a 30-40 day period. 3-4 days after a final boost, draining lymph nodes were harvested. B cells from spleen or lymph node cells were fused with SP2/O-Ag 14 cells (Shulman, Wilde et al. 1978) using conventional techniques. ELISA was used to screen hybridoma supernatants against the receptor ectodomain fusion protein compared to the fusion partner alone, or versus the receptor ectodomain fused to AP (Bates, Fournier et al. 1999). Positive wells were then screened in FACS using 293F cells transiently transfected with expression plasmids encoding full-length receptor cDNAs. Selected hybridomas were single cell cloned and expanded in CELLine flasks (Intergra). Hybridoma supernatants were mixed with an equal volume of 1.5 M glycine, 3 M NaCl, 1×PBS, pH 7.8 and tumbled with MabSelect resin. The resin was washed with binding buffer and eluted with 0.1 M glycine, pH 2.7. Following neutralization with 2 M Tris, mAbs were dialyzed versus PBS.
Characterization of purified anti-DC-ASGPR monoclonal antibodies by direct ELISA. the relative affinities of several anti-DC-ASGPR mAbs by ELISA were determined (i.e., DC-ASGPR.Ig protein is immobilized on the microplate surface and the antibodies are tested in a dose titration series for their ability to bind to DC-ASGPR.Ig (as detected by an anti-mouse IgG.HRP conjugate reagent. In this example, PAB42 and PAB44 show higher affinity binding than other mAbs. The same mAbs fail to bind significantly to human Ig bound to the microplate surface. This shows that the mAbs react to the DC-ASGPR ectodomain part of the DC-ASGPR.Ig fusion protein (data not shown).
Characterization of purified anti-DC-ASGPR monoclonal antibodies by indirect ELISA. Next, the relative affinities of several anti-DC-ASGPR mAbs were determined by ELISA (i.e., anti-DC-ASGPR mAb is immobilized on the microplate surface and tested in a dose titration series for their ability to bind to DC-ASGPR.AP reagent. It was found that the supernatants from the hybridomas listed as: PAB42, PAB44 and PAB54 show higher affinity binding than other mAbs (data not shown).
Characterization of anti-DC-ASGPR mAbs by FACS. The panel of mAbs was also tested by FACS versus 293F cells transfected with expression plasmid directing synthesis of cell surface DC-ASGPR. Mean fluorescence intensity of the signal was subtracted from the analogous signal versus non-transfected 293F cells. By this criterion, the mAbs are able to bind to specifically to the surface of cells bearing DC-ASGPR. Some mAbs, e.g., 37A7 appear particularly advantageous in this regard (data not shown).
FIGS. 1A to 1D shows that signaling through DC-ASGPR activates DCs. DCs are the primary immune cells that determine the results of immune responses, either induction or tolerance, depending on their activation (15). The role of LLRs in DC activation is not clear yet. Therefore, we tested whether triggering the LLR DC-ASGPR can result in the activation of DCs. Both three and six day in vitro cultured GM/IL-4 DCs express LOX-1, ASGPR, and CLEC-6 (FIG. 1A). Six day DCs were stimulated with mAb specific to DC-ASGPR, and data in FIG. 1B show that signals through DC-ASGPR could activate DCs, resulting in the increased expression of CD86 and HLA-DR. Triggering DC-ASGPR on DCs also resulted in the increased production of IL-6, MCP-1, IL-12p40, and IL-8 from DCs (FIG. 1C). Other cytokines and chemokines, TNFa, IP-10, MIP-1a, and IL-10, were also significantly increased (data not shown) by signaling through DC-ASGPR, suggesting that DC-ASGPR can deliver cellular signals to activate DCs. Consistently, DCs stimulated with DC-ASGPR specific mAb expressed increased levels of multiple genes, including co-stimulatory molecules as well as chemokine and cytokine-related genes (FIG. 1D). The possible contribution of LLRs in TLR2 and TLR4-mediated immune cell activation has been described previously (13, 16). We observed that signals through DC-AS GPR could synergize with signal through CD40 for a further activation of DCs (FIG. 1E). This is important because LLRs could serve as co-stimulatory molecules during in vivo DC activation. Taken together, data in FIG. 1 prove that signaling through DC-ASGPR can activate DCs and that DC-ASGPR serves as a co-stimulatory molecule for the activation of DCs. DC-ASGPR engagement during CD40-CD40L interaction results in dramatically increased production of IL-12p70.
DCs stimulated through DC-ASGPR induce potent humoral immune responses. DCs play an important role in humoral immune responses by providing signals for both T-dependent and T-independent B cell responses (19-22) and by transferring antigens to B cells (23, 24). In addition to DCs, signaling through TLR9 as a third signal is necessary for efficient B cell responses (25, 26).
Therefore, we tested the role of DC-ASGPR in DCs-mediated humoral immune responses in the presence of TLR9 ligand, CpG. Six day GM/IL-4 DCs were stimulated with anti-DC-ASGPR mAb, and then purified B cells were co-cultured. As shown in FIG. 2A, DCs activated with anti-DC-ASGPR mAb resulted in remarkably enhanced B cell proliferation (CFSE dilution) and plasma cell differentiation (CD38+CD20), compared to DCs stimulated with control mAb. CD38+CD20 B cells have a typical morphology of plasma cells, but they do not express CD138. The majority of proliferating cells did not express CCR2, CCR4, CCR6, or CCR7. The amounts of total immunoglobulins (Igs) produced were measured by ELISA (FIG. 2B). Consistent with the data in FIG. 2A, B cells cultured with anti-DC-ASGPR-stimulated DCs resulted in significantly increased production of total IgM, IgG, and IgA. In addition to the total Igs, we also observed that DCs activated by triggering DC-ASGPR are more potent than DCs stimulated with control mAb for the production of influenza-virus-specific IgM, IgG, and IgA (FIG. 2C) by B cells, suggesting that DC-ASGPR-mediated DC activation contributes to both total and antigen specific humoral immune responses. We tested the role of DC-ASGPR in ex vivo antigen presenting cells (APCs) in humoral immune responses. Parts of APCs in PBMCs, including CD19+ and CD14+ cells, express DC-ASGPR (Supplementary FIG. 2). PBMCs from buffy coats were cultured in the plates coated with anti-DC-ASGPR mAb, and the total Igs and B cell proliferation were measured. Consistent with the data generated from DCs (FIG. 2A), APCs stimulated through DC-ASGPR resulted in enhanced B cell proliferation and plasma cell differentiation in the absence (upper panels in FIG. 2d ) or presence (lower panels in FIG. 2D) of TLR9 ligand. The total IgM, IgG, and IgA were also significantly increased when PBMCs were cultured in the plates coated with mAb against DC-ASGPR (FIG. 2e ). As shown in FIG. 1, DCs activated by signaling through DC-ASGPR have matured phenotypes and produce large amounts of inflammatory cytokines and chemokines, and both matured DC phenotypes and soluble factors from DCs could contribute to the enhanced B cells responses (FIG. 2). However, DC-derived B lymphocyte stimulator protein (BLyS, BAFF) and a proliferation-inducing ligand (APRIL) are also important molecules by which DCs can directly regulate human B cell proliferation and function (27-30). Therefore, we tested whether signals through DC-ASGPR could alter the expression levels of BLyS and APRIL. Data in FIG. 2d show that DCs stimulated through DC-ASGPR expressed increased levels of intracellular APRIL as well as APRIL secreted, but not BLyS (not shown). Expression levels of BLyS and APRIL receptors on B cells in the mixed cultures were measured, but there was no significant change (not shown).
DC-ASGPR contributes to B cell activation and Ig production. CD19+ B cells express DC-ASGPR (FIG. 3A). Therefore, we tested the role of DC-ASGPR in B cell activation. Data in FIG. 3B show that B cells stimulated through DC-ASGPR produced significantly higher amounts of chemokines. In addition to IL-8 and MIP-1a, slight increases in IL-6 and TNFa were also observed when B cells were stimulated with the anti-DC-ASGPR mAb, compared to control mAb. Genes related to cell activation were also up-regulated (FIG. 3C). B cells produced IgM, IgG, and IgA when they were stimulated through DC-ASGPR (FIG. 3D), suggesting that DC-ASGPR could play an important role in the maintenance of normal immunoglobulin levels in vivo. However, signaling through DC-ASGPR alone did not induce significant B cell proliferation.
Role of DC-ASGPR in T cell responses. DCs stimulated through DC-ASGPR express enhanced levels of co-stimulatory molecules and produce increased amounts of cytokines and chemokines (see FIG. 1), suggesting that DC-ASGPR contributes to cellular immune responses as well as humoral immune responses. This was tested by a mixed lymphocyte reaction (MLR). Proliferation of purified allogeneic T cells was significantly enhanced by DCs stimulated with mAb specific for DC-ASGPR (FIG. 4A). DCs activated through DC-ASGPR could also prime Mart-1-specific CD8 T cells more efficiently than DC stimulated with control mAb (upper panels in FIG. 4B). More importantly, signaling through DC-ASGPR permitted DCs to cross-prime Mart-1 peptides to CD8 T cells (lower panels in FIG. 4B). This indicates that DC-ASGPR plays an important role in enhancing DC function, resulting in better priming and cross-priming of antigens to CD8 T cells. The role of DC-ASGPR expressed on the mixture of APCs in PBMCs in activation of T cell responses is shown in FIG. 4C where PBMCs stimulated with mAb to DC-ASGPR resulted in an increased frequency of Flu M1 tetramer specific CD8 T cells compared to DCs stimulated with control mAb. This enhanced antigen specific CD8 T cell response was supported by the data in FIG. 4D, showing that DCs stimulated through DC-ASGPR significantly increase CD4 T cell proliferation.
Antibodies and tetramers—Antibodies (Abs) for surface staining of DCs and B cells, including isotype control Abs, were purchased from BD Biosciences (CA). Abs for ELISA were purchased from Bethyl (TX). Anti-BLyS and anti-APRIL were from PeproTech (NJ). Tetramers, HLA-A*0201—GILGFVFTL (SEQ ID NO.: 1) (Flu M1) and HLA-A*0201-ELAGIGILTV (SEQ ID NO.: 2) (Mart-1), were purchased from Beckman Coulter (CA).
Cells and cultures—Monocytes (1×106/ml) from normal donors were cultured in Cellgenics (France) media containing GM-CSF (100 ng/ml) and IL-4 (50 ng/ml) (R&D, CA). For day three and day six, DCs, the same amounts of cytokines were supplemented into the media on day one and day three, respectively. B cells were purified with a negative isolation kit (BD). CD4 and CD8 T cells were purified with magnetic beads coated with anti-CD4 or CD8 (Milteniy, Calif.). PBMCs were isolated from Buffy coats using Percoll™ gradients (GE Healthcare UK Ltd, Buckinghamshire, UK) by density gradient centrifugation. For DC activation, 1×105 DCs were cultured in the mAb-coated 96-well plate for 16-18 h. mAbs (1-2 μg/well) in carbonate buffer, pH 9.4, were incubated for at least 3 h at 37° C. Culture supernatants were harvested and cytokines/chemokines were measured by Luminex (Biorad, CA). For gene analysis, DCs were cultured in the plates coated with mAbs for 8 h. In some experiments, soluble 50 ng/ml of CD40L (R&D, CA) or 50 nM CpG (InVivogen, CA) was added into the cultures. In the DCs and B cell co-cultures, 5×103 DCs resuspended in RPMI 1640 with 10% FCS and antibiotics (Biosource, CA) were first cultured in the plates coated with mAbs for at least 6 h, and then 1×105 purified autologous B cells labeled with CFSE (Molecular Probes, OR) were added. In some experiments, DCs were pulsed with 5 moi (multiplicity of infection) of heat-inactivated influenza virus (A/PR/8 H1N1) for 2 h, and then mixed with B cells. For the DCs and T cell co-cultures, 5×103 DCs were cultured with 1×105 purified autologous CD8 T cells or mixed allogeneic T cells. Allogeneic T cells were pulsed with 1 μCi/well 3[H]-thymidine for the final 18 h of incubation, and then cpm were measured by a μ-counter (Wallac, Minn.). 5×105 PBMCs/well were cultured in the plates coated with mAbs. The frequency of Mart-1 and Flu M1 specific CD8 T cells was measured by staining cells with anti-CD8 and tetramers on day ten and day seven of the cultures, respectively. 10 μM of Mart-1 peptide (ELAGIGILTV) (SEQ ID NO.: 2) and 20 nM of recombinant protein containing Mart-1 peptides (see below) were added to the DC and CD8 T cell cultures. 20 nM purified recombinant Flu M1 protein (see below) was add to the PBMC cultures.
Monoclonal antibodies—Mouse mAbs were generated by conventional technology. Briefly, six-week-old BALB/c mice were immunized i.p. with 20 μg of receptor ectodomain.hIgGFc fusion protein with Ribi adjuvant, then boosts with 20 μg antigen ten days and fifteen days later. After three months, the mice were boosted again three days prior to taking the spleens. Alternately, mice were injected in the footpad with 1-10 μg antigen in Ribi adjuvant every three to four days over a thirty to forty day period. Three to four days after a final boost, draining lymph nodes were harvested. B cells from spleen or lymph node cells were fused with SP2/0-Ag 14 cells. Hybridoma supernatants were screened to analyze Abs to the receptor ectodomain fusion protein compared to the fusion partner alone, or the receptor ectodomain fused to alkaline phosphatase (44). Positive wells were then screened in FACS using 293F cells transiently transfected with expression plasmids encoding full-length receptor cDNAs. Selected hybridomas were single cell cloned and expanded in CELLine flasks (Integra, CA). Hybridoma supernatants were mixed with an equal volume of 1.5 M glycine, 3 M NaCl, 1×PBS, pH 7.8 and tumbled with MabSelect resin. The resin was washed with binding buffer and eluted with 0.1 M glycine, pH 2.7. Following neutralization with 2 M Tris, mAbs were dialyzed versus PBS.
ELISA—Sandwich ELISA was performed to measure total IgM, IgG, and IgA as well as flu-specific immunoglobulins (Igs). Standard human serum (Bethyl) containing known amounts of Igs and human AB serum were used as standard for total Igs and flu-specific Igs, respectively. Flu specific Ab titers, units, in samples were defined as dilution factor of AB serum that shows an identical optical density. The amounts of BAFF and BLyS were measured by ELISA kits (Bender MedSystem, CA).
RNA purification and gene analysis—Total RNA extracted with RNeasy columns (Qiagen), and analyzed with the 2100 Bioanalyser (Agilent). Biotin-labeled cRNA targets were prepared using the Illumina totalprep labeling kit (Ambion) and hybridized to Sentrix Human6 BeadChips (46K transcripts). These microarrays consist of 50 mer oligonucleotide probes attached to 3 μm beads which are lodged into microwells etched at the surface of a silicon wafer. After staining with Streptavidin-Cy3, the array surface is imaged using a sub-micron resolution scanner manufactured by Illumina (Beadstation 500X). A gene expression analysis software program, GeneSpring, Version 7.1 (Agilent), was used to perform data analysis.
Expression and purification of recombinant Flu M1 and MART-1 proteins—PCR was used to amplify the ORF of Influenza A/Puerto Rico/8/34/Mount Sinai (H1N1) M1 gene while incorporating an Nhe I site distal to the initiator codon and a Not I site distal to the stop codon. The digested fragment was cloned into pET-28b(+) (Novagen), placing the M1 ORF in-frame with a His6 tag, thus encoding His.Flu M1 protein. A pET28b (+) derivative encoding an N-terminal 169 residue cohesin domain from C. thermocellum (unpublished) inserted between the Nco I and Nhe I sites expressed Coh.His. For expression of Cohesin-Flex-hMART-1-PeptideA-His, the sequence GACACCACCGAGGCCCGCCACCCCCACCCCCCCGTGACCACCCCCACCACCACCGA CCGGAAGGGCACCACCGCCGAGGAGCTGGCCGGCATCGGCATCCTGACCGTGATCC TGGGCGGCAAGCGGACCAACAACAGCACCCCCACCAAGGGCGAATTCTGCAGATA TCCATCACACTGGCGGCCG (SEQ ID NO.: 3) (encoding DTTEARHPHPPVTTPTTDRKGTTAEELAGIGILTVILGGKRTNNSTPTKGEFCRYPSHWR P (SEQ ID NO.: 4)—the marked residues are the immunodominant HLA-A2-restricted peptide and the underlined residues surrounding the peptide are from MART-1) was inserted between the Nhe I and Xho I sites of the above vector. The proteins were expressed in E. coli strain BL21 (DE3) (Novagen) or T7 Express (NEB), grown in LB at 37° C. with selection for kanamycin resistance (40 μg/ml) and shaking at 200 rounds/min to mid log phase growth when 120 mg/L IPTG was added. After three hours, the cells were harvested by centrifugation and stored at −80° C. E. coli cells from each 1 L fermentation were resuspended in 30 ml ice-cold 50 mM Tris, 1 mM EDTA pH 8.0 (buffer B) with 0.1 ml of protease inhibitor Cocktail II (Calbiochem, CA). The cells were sonicated on ice 2×5 min at setting 18 (Fisher Sonic Dismembrator 60) with a 5 min rest period and then spun at 17,000 r.p.m. (Sorvall SA-600) for 20 min at 4° C. For His.Flu M1 purification the 50 ml cell lysate supernatant fraction was passed through 5 ml Q Sepharose beads and 6.25 ml 160 mM Tris, 40 mM imidazole, 4 M NaCl pH 7.9 was added to the Q Sepharose flow through. This was loaded at 4 ml/min onto a 5 ml HiTrap chelating HP column charged with Ni++. The column-bound protein was washed with 20 mM NaPO4, 300 mM NaCl pH 7.6 (buffer D) followed by another wash with 100 mM H3COONa pH 4.0. Bound protein was eluted with 100 mM H3COONa pH 4.0. The peak fractions were pooled and loaded at 4 ml/min onto a 5 ml HiTrap S column equilibrated with 100 mM H3COONa pH 5.5, and washed with the equilibration buffer followed by elution with a gradient from 0-1 M NaCl in 50 mM NaPO4 pH 5.5. Peak fractions eluting at about 500 mM NaCl were pooled. For Coh.Flu M1.His purification, cells from 2 L of culture were lysed as above. After centrifugation, 2.5 ml of Triton X114 was added to the supernatant with incubation on ice for 5 min. After further incubation at 25° C. for 5 min, the supernatant was separated from the Triton X114 following centrifugation at 25° C. The extraction was repeated and the supernatant was passed through 5 ml of Q Sepharose beads and 6.25 ml 160 mM Tris, 40 mM imidazole, 4 M NaCl pH 7.9 was added to the Q Sepharose flow through. The protein was then purified by Ni++ chelating chromatography as described above and eluted with 0-500 mM imidazole in buffer D.
Only particular anti-DC-ASGPR mAbs have DC activation properties—The invention discloses that DC activation is not a general property of anti-DC-ASGPR antibodies, rather only certain anti-DC-ASGPR mAbs have this function. FIG. 5 shows that only certain mAbs activate DCS through the DC-ASGPR, which must be characterized by screening against actual DCs.
Particular sequences corresponding to the L and H variable regions of anti-DC-ASGPR mAbs—The invention encompasses particular amino acid sequences shown below corresponding to anti-DC-ASGPR monoclonal antibodies that are desirable components (in the context of e.g., humanized recombinant antibodies) of therapeutic or protective products. The following are such sequences in the context of chimeric mouse V region—human C region recombinant antibodies. [mAnti-ASGPR_49C11_7H-LV-hIgG4H-C] is
(SEQ ID NO.: 5)
DVQLQESGPDLVKPSQSLSLTCTVTGYSITSGYSWHWIRQFPGNKLEW
MGYILFSGSTNYNPSLKSRISITRDTSKNQFFLQLNSVTTEDTATYFC
ARSNYGSFASWGQGTLVTVSAAKTTGPSVFPLAPCSRSTSESTAALGC
LVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSS
LGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEFEGGPSVFLF
PPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKP
REEQFNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKA
KGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQP
ENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEALHNH
YTQKSLSLSLGKAS.

The above sequence is a chimera between the H chain V-region of the mAb 49C11 (shown underlined) and the C region of hIgG4. [mAnti-ASGPR_49C11_7K-LV-hIgGK-C] is the corresponding L chain chimera
[mAnti-ASGPR_4G2.2_Hv-V-hIgG4H-C]
(SEQ ID NO.: 6)
-QIVLTQSPAIMSASPGEKVTMTCSASSSVSHMHWYQQKSGTSPKRWIY
DTSRLASGVPARFSGSGSGTSYSLTISSMEAEDAATYYCQQWSSHPWS
FGGGTKLEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAK
VQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYA
CEVTHQGLSSPVTKSFNRGEC.
is
[mAnti-ASGPR_4G2.2_Kv-V-hIgGK-C]
(SEQ ID NO.: 7)
-QIQLVQSGPELKKPGETVKISCKASGYTFTNYGMNWVKQVPGKGLRWM
GWMDTFTGEPTYADDFKGRFAFSLETSASTAYLQINSLKNEDTATYFC
ARGGILRLNYFDYWGQGTTLTVSSAKTKGPSVFPLAPCSRSTSESTAA
LGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVP
SSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEFEGGPSV
FLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAK
TKPREEQFNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTI
SKAKGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESN
GQPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEAL
HNHYTQKSLSLSLGKAS.
is
[mAnti-ASGPR_5F10H-LV-hIgG4H-C]
(SEQ ID NO.: 8)
-DIQMTQSSSSFSVSLGDRVTITCKASEDIYNRLGWYQQKPGNAPRLLI
SGATSLETGVPSRFSGSGSGKDYALSITSLQTEDLATYYCQQCWTSPY
TFGGGTKLEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREA
KVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVY
ACEVTHQGLSSPVTKSFNRGEC.
is
[mAnti-ASGPR_5F10K-LV-hIgGK-C]
(SEQ ID NO.: 9)
-EVQLQQSGPELVKPGASVKMSCKASGYTFTDYYMKWVKQSHGKSLEWI
GDINPNYGDTFYNQKFEGKATLTVDKSSRTAYMQLNSLTSEDSAVYYC
GRGDYGYFDVWGAGTTVTVSSAKTKGPSVFPLAPCSRSTSESTAALGC
LVKDYFPEPVTVSWNGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSL
GTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEFEGGPSVFLFP
PKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPR
EEQFNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAK
GQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPE
NNYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEALHNHY
TQKSLLSLGKAS.
is
[mAnti-ASGPR1H11H-V-hIgG4H-C]
(SEQ ID NO.: 10)
-DIVMTQSHKFMSTSVGDRVSITCKASQDVGTAVAWYQQKPGQSPKLLI
YWASTRHTGVPDRFTGSGSGTDFTLTINNVQSEDLADYFCQQYSSNPY
MFGGGTKLEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREA
KVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVY
ACEVTHQGLSSPVTKSFNRGEC.
is
[mAnti-ASGPR1H11K-LV-hIgGK-C]
(SEQ ID NO.: 11)
-QLQQSGPELVKPGASVKISCKTSGYTFTEYTMHWVRQSHGKSLEWIGG
INPINGGPTYNQKFKGKATLTVDKSSSTAYMELRSLTSEDSAVYYCAR
WDYGSRDVMDYWGQGTSVTVSSAKTKGPSVFPLAPCSRSTSESTAALG
CLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSS
SLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEFEGGPSVFL
FPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTK
PREEQFNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISK
AKGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQ
PENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEALHN
HYTQKSLSLSLGKAS.
is
(SEQ ID NO.: 12)
-NIVMTQSPKSMSMSVGERVTLSCKASENVGTYVSWYQQRPEQSPKLLI
YGASNRYTGVPDRFTGSGSATDFTLTISSVQAEDLADYHCGQTYSYIF
TFGSGTKLEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREA
KVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVY
ACEVTHQGLSSPVTKSFNRGEC.

The invention envisions these V-region sequences and related sequences modified by those well versed in the art to e.g., enhance affinity for DC-ASGPR and/or integrated into human V-region framework sequences to be engineered into expression vectors to direct the expression of protein forms that can bind to DC-ASGPR on antigen presenting cells.
Engineered recombinant anti-DC-ASGPR recombinant antibody—antigen fusion proteins ((rAb.antigen) are efficacious prototype vaccines in vitro—Expression vectors can be constructed with diverse protein coding sequence e.g., fused in-frame to the H chain coding sequence. For example, antigens such as Influenza HAS, Influenza M1, HIV gag, or immuno-dominant peptides from cancer antigens, or cytokines, can be expressed subsequently as rAb.antigen or rAb.cytokine fusion proteins, which in the context of this invention, can have utility derived from using the anti-DC-ASGPR V-region sequence to bring the antigen or cytokine (or toxin) directly to the surface of the antigen presenting cell bearing DC-AS GPR. This permits internalization of e.g., antigen—sometimes associated with activation of the receptor and ensuing initiation of therapeutic or protective action (e.g., via initiation of a potent immune response, or via killing of the targeted cell. An exemplative prototype vaccine based on this concept could use a H chain vector such as [mAnti-ASGPR_5F10H-LV-hIgG4H-C-Flex-FluHA5-1-6×His] or
(SEQ ID NO.: 13)
EVQLQQSGPELVKPGASVKMSCKASGYTFTDYYMKWVKQSHGKSLEWI
GDINPNYGDTFYNQKFEGKATLTVDKSSRTAYMQLNSLTSEDSAVYYC
GRGDYGYFDVWGAGTTVTVSSAKTKGPSVFPLAPCSRSTSESTAALGC
LVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSS
LGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEFEGGPSVFLF
PPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKP
REEQFNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKA
KGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQP
ENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEALHNH
YTQKSLSLSLGKASDTTEPATPTTPVTT DQICIGYHANNSTEQVDTIM
EKNVTVTHAQDILEKKHNGKLCDLDGVKPLILRDCSVAGWLLGNPMCD
EFINVPEWSYIVEKANPVNDLCYPGDFNDYEELKHLLSRINHFEKIQI
IPKSSWSSHEASLGVSSACPYQGKSSFFRNVVWLIKKNSTYPTIKRSY
NNTNQEDLLVLWGIHHPNDAAEQTKLYQNPTTYISVGTSTLNQRLVPR
IATRSKVNGQSGRMEFFWTILKPNDAINFESNGNFIAPEYAYKIVKKG
DSTIMKSELEYGNCNTKCQTPMGAINSSMPFHNIHPLTIGECPKYVKS
NRLVLAHHHHHH.

The above sequence corresponds to the chimeric H chain shown already fused via a flexible linker sequence (shown italicized) to HA-1 domain of avian Flu HAS (shown in bold). This can be co-expressed with the corresponding L chain chimeric sequence already shown above. Similarly, the sequence [mAnti-ASGPR_49C11_7H-LV-hIgG4H-C-Dockerin]
(SEQ ID NO.: 14)
DVQLQESGPDLVKPSQSLSLTCTVTGYSITSGYSWHWIRQFPGNKLEW
MGYILFSGSTNYNPSLKSRISITRDTSKNQFFLQLNSVTTEDTATYFC
ARSNYGSFASWGQGTLVTVSAAKTKGPSVFPLAPCSRSTSESTAALGC
LVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSS
LGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEFEGGPSVFLF
PPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKP
REEQFNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKA
KGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQP
ENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEALHNH
YTQKSLSLSLGKASNSPQNEVLYGDVNDDGKVNSTDLTLLKRYVLKAV
STLPSSKAEKNADVNRDGRVNSSDVTILSRYLIRVIEKLPI

can be used to express via co-transfection of the corresponding L chain sequence already shown above a rAb.Dockerin fusion protein.
FIG. 5 shows that certain anti-DC-ASGPR mAbs can activate DC. GM-CSF/IL-4. DC were incubated for 24 hrs with one of a panel of 12 pure anti-ASGPR mAbs. Cells were then tested for expression of cell surface CD86 (a DC activation marker) and supernatants were assayed for secreted cytokines. Three mAbs (36, 38, 43) from the anti-ASGPR mAb panel activated DC.
FIG. 6 shows that different antigens can be expressed in the context of a DC-ASGPR rAb. Such an anti-DC-ASGPR rAb.Doc protein can be simply mixed with any Cohesin.fusion protein to assemble a stable non-covalent [rAb.Doc:Coh.fusion] complex that functions just as a rAb.fusion protein. FIG. 6 shows that such a [rAb.Doc:Coh.fusion] complex can focus antigen to the surface of cells expressing DC-ASGPR. The figure also shows anti-DC-ASGPR.Doc:Coh.Flu M1 complexes deliver Flu M1 to the surface of 293F cells transfected with DC-ASGPR cDNA. 1 μg/ml of anti-DC-ASGPR.Doc rAb (shown as the peak on the right) or control hIgG4.Doc rAb (shown as the peak on the left) were incubated with biotinylated Coh.Flu M1 (2 μg/ml) for 1 hr at R.T. transfected 293F cells were added and incubation continued for 20 min on ice. Cells were then washed and stained with PE-labeled streptavidin. Cells were then analyzed for PE fluorescence.
Anti-DC-ASGPR rAb complexed to Flu M1 via Dockerin:Cohesin interaction targets the antigen to human DCs and results in the expansion of Flu M1-specific CD8+ T cells—the potential utility of anti-DC-ASGPR rAbs as devices to deliver antigen to e.g., DC is shown in the figure below. FIG. 7 shows the dramatic expansion of Flu M1-specific CD8+ cells is highly predictive of potency of such an agent as a vaccine directed to eliciting protective immune responses against Flu M1.
FIGS. 8A-8D demonstrated the cross reactivity of the different antibodies with monkey ASGPR. For pIRES_ ASGPR-mon (monkey) was cloned by inserting the PCR product into NheI-NotI sites of pIRES vector. The sequence of final product is base on clone 5S10. Most other clones are either similar to this with one aa difference or identical to this. However, one clone, 5S1, has an A deletion near the 3′ end, which generated a shortened and different C′ terminus and maybe used as a second variant. To clone the monkey ASGPR, the following oligos were used: DC-ASGPR_MoN: gaattcgctagcCACCATGACATATGAAAACTTCCAAGACTTGGAGAGTGAGGAGAAAGT CCAAGGGG (SEQ ID NO.: 15); and DC-ASGPR_Mo: CGAATTCGCGGCCGCTCAGTGACTCTCCTGGCTGGCCTGGGTCAGACCAGCCTCGC AGACCC (SEQ ID NO.: 16), which is a reverse complement of GGGTCTGCGAGGCTGGTCTGACCCAGGCCAGCCAGGAGAGTCACTGAGCGGCCGC GAATTCG (SEQ ID NO.: 17). Sequence comparisons indicate the likely regions of overlap and, hence, the cross-reactivity, as is known to those if skill in the art.
The following table demonstrated the binding of the DC-ASGPR 334998 200 ug/ml 12.05.07 cfg#558 anti-Human IgG PE
Avg StDev SEM
w/o w/o w/o
Glycan Max & Max & Max &
number Glycan name Min Min Min % CV
82 GalNAcα1-3(Fucα1-2)Galβ1-4GlcNAcβ-Sp8 52930 10265 5132 19
210 Neu5Acα2-3(GalNAcβ1-4)Galβ1-4GlcNAcβ-Sp8 49937 4969 2484 10
86 GalNAcα1-3Galβ-Sp8 49067 4672 2336 10
89 GalNAcβ1-3(Fucα1-2)Galβ-Sp8 47375 5453 2726 12
84 GalNAcα1-3(Fucα1-2)Galβ-Sp8 46555 6618 3309 14
209 Neu5Acα2-3(GalNAcβ1-4)Galβ1-4GlcNAcβ-Sp0 46169 2121 1060 5
175 GlcNAcβ1-6GalNAcα-Sp8 44809 1939 969 4
301 GalNAcα1-3(Fucα1-2)Galβ-Sp18 44147 6003 3002 14
211 Neu5Acα2-3(GalNAcβ1-4)Galβ1-4Glcβ-Sp0 43603 3517 1759 8
10 α-GalNAc-Sp8 43514 2476 1238 6
128 Galβ1-3GalNAcβ1-4(Neu5Acα2-3)Galβ1-4Glcβ-Sp0 43152 13339 6669 31
151 Galβ1-4GlcNAcβ1-6GalNAcα-Sp8 42871 2466 1233 6
92 GalNAcβ1-4GlcNAcβ-Sp0 42845 3394 1697 8
93 GalNAcβ1-4GlcNAcβ-Sp8 41764 7340 3670 18
87 GalNAcα1-4(Fucα1-2)Galβ1-4GlcNAcβ-Sp8 41584 2925 1462 7
79 GalNAcα1-3(Fucα1-2)Galβ1-3GlcNAcβ-Sp0 41406 14134 7067 34
20 β-GalNAc-Sp8 40803 2388 1194 6
206 Neu5Acα2-8Neu5Acα2-3(GalNAcβ1-4)Galβ1-4Glcβ-Sp0 38720 2736 1368 7
242 Neu5Acα2-6GalNAcα-Sp8 37500 1934 967 5
91 GalNAcβ1-4(Fucα1-3)GlcNAcβ-Sp0 37286 5046 2523 14
204 Neu5Acα2-8Neu5Acα2-8Neu5Acα2-3(GalNAcβ1-4)Galβ1- 37237 995 497 3
4Glcβ-Sp0
203 NeuAcα2-8NeuAcα2-8NeuAcα2-8NeuAcα2-3(GalNAcβ1-4) 36746 2399 1200 7
Galβ1-4Glcβ-Sp0
243 Neu5Acα2-6GalNAcβ1-4GlcNAcβ-Sp0 36375 1661 830 5
59 Fucα1-2Galβ1-3GalNAcβ1-4(Neu5Acα2-3)Galβ1-4Glcβ-Sp0 35701 6903 3452 19
90 GalNAcβ1-3Galα1-4Galβ1-4GlcNAcβ-Sp0 34350 760 380 2
83 GalNAcα1-3(Fucα1-2)Galβ1-4Glcβ-Sp0 28846 9844 4922 34
302 GalNAcβ1-3Galβ-Sp8 28745 15727 7864 55
300 GalNAcα-Sp15 18125 18847 9424 104
127 Galβ1-3GalNAcβ1-3Galα1-4Galβ1-4Glcβ-Sp0 17999 9798 4899 54
85 GalNAcα1-3GalNAcβ-Sp8 12643 10843 5422 86
173 GlcNAcβ1-4GlcNAcβ1-4GlcNAcβ-Sp8 8673 940 470 11
81 GalNAcα1-3(Fucα1-2)Galβ1-4GlcNAcβ-Sp0 7672 12937 6469 169
30 [3OSO3]Galβ1-4(6OSO3)Glcβ-Sp8 7394 292 146 4
120 Galβ1-3(Galβ1-4GlcNAcβ1-6)GalNAcα-Sp8 5664 1311 655 23
80 GalNAcα1-3(Fucα1-2)Galβ1-4(Fucα1-3)GlcNAcβ-Sp0 5444 907 454 17
147 Galβ1-4GlcNAcβ1-3Galβ1-4GlcNAcβ-Sp0 4927 410 205 8
29 [3OSO3]Galβ1-4(6OSO3)Glcβ-Sp0 4871 908 454 19
101 Galα1-3GalNAcα-Sp8 4815 3163 1581 66
214 Neu5Acα2-3GalNAcα-Sp8 4109 569 284 14
287 [3OSO3][4OSO3]Galβ1-4GlcNacβ-SpSp0 3959 1646 823 42
40 [4OSO3]Galβ1-4GlcNAcβ-Sp8 3848 673 337 17
45 [6OSO3]Galβ1-4[6OSO3]Glcβ-Sp8 3790 993 497 26
166 GlcNAcβ1-3Galβ1-4GlcNAcβ1-3Galβ1-4GlcNAcβ-Sp0 3720 435 218 12
227 Neu5Acα2-3Galβ1-4[6OSO3]GlcNAcβ-Sp8 3576 793 397 22
218 NeuAcα2-3Galβ1-3(Fucα1-4)GlcNAcβ1-3Galβ1-4(Fucα1-3) 3360 104 52 3
GlcNAcβ Sp0
240 Neu5Acα2-3Galβ1-4Glcβ-Sp8 3313 976 488 29
149 Galβ1-4GlcNAcβ1-3Galβ1-4Glcβ-Sp8 3233 263 132 8
244 Neu5Acα2-6Galβ1-4[6OSO3]GlcNAcβ-Sp8 3195 757 379 24
270 Fucα1-2Galβ1-4[6OSO3]GlcNAc-Sp8 3161 2563 1282 81
42 [6OSO3]Galβ1-4Glcβ-Sp0 3084 529 264 17
271 Fucα1-2[6OSO3]Galβ1-4[6OSO3]Glc-Sp0 3063 377 188 12
172 (GlcNAcβ1-4)5β-Sp8 3032 1058 529 35
47 [6OSO3]GlcNAcβ-Sp8 3008 159 80 5
143 Neu5Acα2-3Galβ1-4GlcNAcβ1-2Manα1-3(Neu5Acα2- 3008 309 155 10
3Galβ1-4GlcNAcβ1-2Manα1-6)Manβ1-4GlcNAcβ1-
4GlcNAcβ-Sp12
265 [3OSO3]Galβ1-4(Fucα1-3)(6OSO3)Glc-Sp0 2995 1841 921 61
139 Galβ1-4[6OSO3]Glcβ-Sp0 2988 1070 535 36
27 [3OSO3][6OSO3]Galβ1-4GlcNAcβ-Sp0 2930 317 158 11
273 Fucα1-2-Galβ1-4[6OSO3]Glc-Sp0 2919 495 247 17
319 Neu5Acα2-6Galβ1-4GlcNAcβ1-2Manα1-3(Galβ1- 2730 993 497 36
4GlcNAcβ1-2Manα1-6)Manβ1-4GlcNAcβ1-
4GlcNAcβ1-Sp12
35 [3OSO3]Galβ1-4[6OSO3]GlcNAcβ-Sp8 2722 516 258 19
28 [3OSO3]Galβ1-4Glcβ-Sp8 2674 197 98 7
38 [3OSO3]Galβ-Sp8 2652 1680 840 63
253 Neu5Acα2-8Neu5Acα2-3Galβ1-4Glcβ-Sp0 2631 1136 568 43
289 6-H2PO3Glcβ-Sp10 2611 674 337 26
26 [3OSO3][6OSO3]Galβ1-4[6OSO3]GlcNAβ-Sp0 2550 153 76 6
266 [3OSO3]Galβ1-4(Fucα1-3)Glc-Sp0 2529 444 222 18
54 Neu5Acα2-6Galβ1-4GlcNAcβ1-2Manα1-3(Neu5Acα2- 2476 300 150 12
6Galβ1-4GlcNAcβ1-2Manα1-6)Manβ1-4GlcNAcβ1-
4GlcNAcβ-Sp8
303 GlcAβ1-3GlcNAcβ-Sp8 2463 130 65 5
32 [3OSO3]Galβ1-3GalNAcα-Sp8 2461 622 311 25
53 Neu5Acα2-6Galβ1-4GlcNAcβ1-2Manα1-3(Neu5Acα2- 2455 283 142 12
6Galβ1-4GlcNAcβ1-2Manα1-6)Manβ1-4GlcNAcβ1-
4GlcNAcβ-Sp13
181 Glcβ1-6Glcβ-Sp8 2455 154 77 6
267 [3OSO3]Galβ1-4[Fucα1-3][6OSO3]GlcNAc-Sp8 2447 1065 532 44
293 Galβ1-3(Neu5Acα2-3Galβ1-4(Fucα1-3)GlcNAcβ1-6) 2359 648 324 27
GalNAc-Sp14
202 Neu5Acα2-3Galβ1-3GalNAcα-Sp8 2349 928 464 40
163 GlcNAcβ1-3Galβ1-3GalNAcα-Sp8 2347 375 188 16
1 Neu5Acα2-8Neu5Acα-Sp8 2339 1539 769 66
31 [3OSO3]Galβ1-3(Fucα1-4)GlcNAcβ-Sp8 2332 319 160 14
230 Neu5Acα2-3Galβ1-4(Fucα1-3)GlcNAcβ-Sp0 2306 164 82 7
286 [3OSO3]Galβ1-4[6OSO3]GlcNAcβ-Sp0 2290 472 236 21
318 Neu5Acα2-3Galβ1-4GlcNAcβ1-2Manα1-3(Neu5Acα2- 2262 246 123 11
6Galβ1-4GlcNAcβ1-2Manα1-6)Manβ1-4GlcNAcβ1-
4GlcNAcβ-Sp12
199 Neu5Acα2-6Galβ1-4GlcNAcβ1-2Manα1-3(Neu5Acα2- 2217 138 69 6
3Galβ1-4GlcNAcβ1-2Manα1-6)Manβ1-4GlcNAcβ1-
4GlcNAcβ-Sp12
39 [4OSO3][6OSO3]Galβ1-4GlcNAcβ-Sp0 2215 619 310 28
77 Fucα1-4GlcNAcβ-Sp8 2207 83 42 4
285 Neu5Acα2-3Galβ1-4GlcNAcβ1-3Galβ1-3GlcNAcβ-Sp0 2193 1679 839 77
262 Neu5Gcα2-6GalNAcα-Sp0 2192 734 367 33
216 Neu5Acα2-3Galβ1-3(6OSO3)GlcNAc-Sp8 2163 1062 531 49
43 [6OSO3]Galβ1-4Glcβ-Sp8 2149 700 350 33
297 Galβ1-4GlcNAcβ1-3(GlcNAcβ1-6)Galβ1-4GlcNAc-Sp0 2141 983 491 46
224 NeuAcα2-3Galβ1-3GlcNAcβ1-3Galβ1-4GlcNAcβ-Sp0 2133 1208 604 57
3 Neu5Acα2-8Neu5Acα2-8Neu5Acβ-Sp8 2117 611 306 29
171 (GlcNAcβ1-4)6β-Sp8 2112 302 151 14
316 Neu5Acα2-3Galβ1-3(Neu5Acα2-6)GalNAc-Sp14 2105 1171 585 56
15 α-Neu5Ac-Sp11 2099 250 125 12
52 Galβ1-4GlcNAcβ1-2Manα1-3(Galβ1-4GlcNAcβ1-2Manα1-6) 2092 429 215 21
Manβ1-4GlcNAcβ1-4GlcNAcβ-Sp13
268 [3OSO3]Galβ1-4[Fucα1-3]GlcNAc-Sp0 2085 955 477 46
313 Manα1-2Manα1-2Manα1-3(Manα1-2Manα1-6(Manα1-3) 2020 812 406 40
Manα1-6)Manα-Sp9
225 Neu5Acα2-3Galβ1-3GlcNAcβ-Sp0 2019 1052 526 52
36 [3OSO3]Galβ1-4GlcNAcβ-Sp0 2012 389 194 19
263 Neu5Gcα2-6Galβ1-4GlcNAcβ-Sp0 1999 664 332 33
141 Galβ1-4GalNAcα1-3(Fucα1-2)Galβ1-4GlcNAcβ-Sp8 1968 772 386 39
274 Galβ1-3(Fucα1-4)GlcNAcβ1-3Galβ1-3(Fucα1-4)GlcNAcβ- 1961 78 39 4
Sp0
275 Galβ1-3-(Galβ1-4GlcNacβ1-6)GalNAc-Sp14 1953 409 205 21
7 α-D-Gal-Sp8 1925 636 318 33
41 6-H2PO3Manα-Sp8 1919 223 111 12
247 Neu5Acα2-6Galβ1-4GlcNAcβ1-3Galβ1-4(Fucα1-3) 1914 169 85 9
GlcNAcβ1-3Galβ1-4(Fucα1-3)GlcNAcβ-Sp0
311 Manα1-6Manβ-Sp10 1906 522 261 27
205 Neu5Acα2-8Neu5Acα2-8Neu5Acα2-3Galβ1-4Glcβ-Sp0 1902 222 111 12
280 Galβ1-4[Fucα1-3][6OSO3]GlcNAc-Sp0 1881 982 491 52
152 Galβ1-4GlcNAcβ-Sp0 1868 924 462 49
113 Galα1-6Glcβ-Sp8 1864 321 161 17
115 Galβ1-3(Fucα1-4)GlcNAcβ1-3Galβ1-4(Fucα1-3)GlcNAcβ- 1855 338 169 18
Sp0
251 Neu5Acα2-6Galβ-Sp8 1842 316 158 17
116 Galβ1-3(Fucα1-4)GlcNAcβ1-3Galβ1-4GlcNAcβ-Sp0 1836 798 399 43
194 Manα1-2Manα1-2Manα1-3(Manα1-2Manα1-3(Manα1- 1829 176 88 10
2Manα1-6)Manα1-6)Manβ1-4GlcNAcβ1-4GlcNAcβ-Sp12
33 [3OSO3]Galβ1-3GlcNAcβ-Sp8 1812 889 445 49
272 Fucα1-2-(6OSO3)-Galβ1-4Glc-Sp0 1805 86 43 5
207 Neu5Acα2-8Neu5Acα2-8Neu5Acα-Sp8 1804 454 227 25
74 Fucα1-2Galβ-Sp8 1796 648 324 36
213 Neu5Acα2-3(Neu5Acα2-6)GalNAcα-Sp8 1768 312 156 18
234 Neu5Acα2-3Galβ1-4GlcNAcβ1-3Galβ1-4(Fucα1-3)GlcNAc- 1767 178 89 10
Sp0
50 Manα1-3(Manα1-6)Manβ1-4GlcNAcβ1-4GlcNAcβ-Sp13 1759 553 277 31
111 Galα1-4Galβ1-4Glcβ-Sp0 1740 635 318 36
291 Galα1-3GalNAcα-Sp16 1738 1090 545 63
296 Galβ1-4GlcNAcβ1-3(Galβ1-4GlcNAcβ1-6)Galβ1-4GlcNAc- 1726 850 425 49
Sp0
154 Galβ1-4Glcβ-Sp0 1725 457 229 27
56 Fucα1-2Galβ1-3GalNAcβ1-3Galα1-4Galβ1-4Glcβ-Sp9 1719 384 192 22
66 Fucα1-2Galβ1-4(Fucα1-3)GlcNAcβ1-3Galβ1-4(Fucα1-3) 1703 224 112 13
GlcNAcβ1-3Galβ1-4(Fucα1-3)GlcNAcβ-Sp0
299 Galβ1-4GlcNAcβ1-6Galβ1-4GlcNAcβ-Sp0 1658 820 410 49
44 [6OSO3]Galβ1-4GlcNAcβ-Sp8 1632 242 121 15
237 Neu5Acα2-3Galβ1-4GlcNAcβ-Sp8 1632 1049 524 64
233 Neu5Acα2-3Galβ1-4(Fucα1-3)GlcNAcβ1-3Galβ1- 1620 862 431 53
4GlcNAcβ-Sp8
192 Manα1-6(Manα1-2Manα1-3)Manα1-6(Manα2Manα1-3) 1608 903 452 56
Manβ1-4GlcNAcβ1-4GlcNAcβ-Sp12
64 Fucα1-2Galβ1-3GlcNAcβ-Sp8 1602 625 313 39
62 Fucα1-2Galβ1-3GlcNAcβ1-3Galβ1-4Glcβ-Sp8 1580 417 208 26
148 Galβ1-4GlcNAcβ1-3Galβ1-4Glcβ-Sp0 1568 617 308 39
295 Galβ1-4GlcNAcβ1-2Manα1-3(Neu5Acα2-6Galβ1- 1556 190 95 12
4GlcNAcβ1-2Manα1-6)Manβ1-4GlcNAcβ1-4GlcNAcβ-Sp12
137 Galβ1-4(Fucα1-3)GlcNAcβ1-4Galβ1-4(Fucα1-3)GlcNAcβ- 1552 1313 656 85
Sp0
17 β-D-Gal-Sp8 1544 871 435 56
168 GlcNAcβ1-4MDPLys 1542 345 172 22
254 Neu5Acβ2-6GalNAcα-Sp8 1541 688 344 45
231 Neu5Acα2-3Galβ1-4(Fucα1-3)GlcNAcβ-Sp8 1534 257 129 17
125 Galβ1-3GalNAcα-Sp8 1483 1025 512 69
269 Fucα1-2[6OSO3]Galβ1-4GlcNAc-Sp0 1473 191 96 13
182 G-ol-Sp8 1471 264 132 18
37 [3OSO3]Galβ1-4GlcNAcβ-Sp8 1462 1187 593 81
229 Neu5Acα2-3Galβ1-4(Fucα1-3)GlcNAcβ1-3Galβ1-4(Fucα1-3) 1451 333 167 23
GlcNAcβ1-3Galβ1-4(Fucα1-3)GlcNAcβ-Sp0
315 Neu5Acα2-3Galβ1-3(Neu5Acα2-3Galβ1-4GlcNAcβ1-6) 1448 1476 738 102
GalNAc-Sp14
65 Fucα1-2Galβ1-4(Fucα1-3)GlcNAcβ1-3Galβ1-4(Fucα1-3) 1442 748 374 52
GlcNAcβ-Sp0
164 GlcNAcβ1-3Galβ1-4GlcNAcβ-Sp0 1436 1332 666 93
305 GlcNAcβ1-2Manα1-3(GlcNAcβ1-2Manα1-6)Manβ1- 1428 288 144 20
4GlcNAcβ1-4GlcNAcβ-Sp12
304 GlcNAcβ1-2Manα1-3(Neu5Acα2-6Galβ1-4GlcNAcβ1- 1428 499 249 35
2Manα1-6)Manβ1-4GlcNAcβ1-4GlcNAcβ-Sp12
145 Galβ1-4GlcNAcβ1-3Galβ1-4(Fucα1-3)GlcNAcβ1-3Galβ1- 1422 323 162 23
4(Fucα1-3)GlcNAcβ-Sp0
117 Galβ1-3(Fucα1-4)GlcNAc-Sp0 1407 681 341 48
193 Manα1-2Manα1-6(Manα1-3)Manα1-6(Manα2Manα2Manα1- 1404 285 142 20
3)Manβ1-4GlcNAcβ1-4GlcNAcβ-Sp12
19 β-D-Man-Sp8 1389 635 317 46
176 GlcNAcβ1-6Galβ1-4GlcNAcβ-Sp8 1383 1000 500 72
232 Neu5Acα2-3Galβ1-4(Fucα1-3)GlcNAcβ1-3Galβ-Sp8 1355 374 187 28
219 Neu5Acα2-3Galβ1-3(Neu5Acα2-3Galβ1-4)GlcNAcβ-Sp8 1350 753 377 56
123 Galβ1-3(Neu5Acβ2-6)GalNAcα-Sp8 1350 852 426 63
276 Galβ1-3(GlcNacβ1-6)GalNAc-Sp14 1345 353 176 26
208 Neu5Acα2-3(6-O-Su)Galβ1-4(Fucα1-3)GlcNAcβ-Sp8 1341 642 321 48
55 Fucα1-2Galβ1-3GalNAcβ1-3Galα-Sp9 1331 466 233 35
257 Neu5Gca2-3Galβ1-3(Fucα1-4)GlcNAcβ-Sp0 1315 108 54 8
201 Fucα1-3(Galβ1-4)GlcNAcβ1-2Manα1-3(Fucα1-3(Galβ1-4) 1294 289 144 22
GlcNAcβ1-2Manα1-6)Manβ1-4GlcNAcβ1-4GlcNAcβ-
Sp20
97 Galα1-3(Fucα1-2)Galβ1-4GlcNAc-Sp0 1282 583 291 45
150 Galβ1-4GlcNAcβ1-6(Galβ1-3)GalNAcα-Sp8 1265 778 389 62
60 Fucα1-2Galβ1-3GalNAcβ1-4(Neu5Acα2-3)Galβ1-4Glcβ-Sp9 1261 738 369 59
317 Neu5Acα2-3Galβ1-3GalNAc-Sp14 1239 780 390 63
23 β-GlcN(Gc)-Sp8 1219 436 218 36
279 Galβ1-3GlcNAcβ1-3Galβ1-3GlcNAcβ-Sp0 1219 570 285 47
190 Manα1-2Manα1-3(Manα1-2Manα1-6)Manα-Sp9 1217 1305 653 107
178 Glcα1-4Glcα-Sp8 1216 560 280 46
146 Galβ1-4GlcNAcβ1-3Galβ1-4GlcNAcβ1-3Galβ1-4GlcNAcβ- 1211 1315 658 109
Sp0
292 Galβ1-3GalNAcα-Sp16 1198 370 185 31
221 Neu5Acα2-3Galβ1-3(Neu5Acα2-6)GalNAcα-Sp8 1194 238 119 20
99 Galα1-3(Fucα1-2)Galβ-Sp8 1189 767 383 64
309 HOOC(CH3)CH-3-O-GlcNAcβ1-4GlcNAcβ-Sp10 1186 1108 554 93
248 Neu5Acα2-6Galβ1-4GlcNAcβ1-3Galβ1-4GlcNAcβ-Sp0 1181 334 167 28
107 Galα1-3Galβ-Sp8 1148 688 344 60
236 Neu5Acα2-3Galβ1-4GlcNAcβ-Sp0 1148 441 220 38
320 Neu5Acα2-6Galβ1-4GlcNAcβ1-2Manα1-3(GlcNAcβ1- 1142 55 27 5
2Manα1-6)Manβ1-4GlcNAcβ1-4GlcNAcβ-Sp12
197 Manα1-6(Manα1-3)Manα1-6(Manα2Manα1-3)Manβ1- 1134 200 100 18
4GlcNAcβ1-4GlcNAcβ-Sp12
185 GlcAβ1-3Galβ-Sp8 1133 470 235 42
34 [3OSO3]Galβ1-4(Fucα1-3)GlcNAcβ-Sp8 1117 980 490 88
109 Galα1-4Galβ1-4GlcNAcβ-Sp0 1094 499 250 46
235 Neu5Acα2-3Galβ1-4GlcNAcβ1-3Galβ1-4GlcNAcβ1-3Galβ1- 1092 1077 539 99
4GlcNAcβ-Sp0
228 Neu5Acα2-3Galβ1-4(Fucα1-3)(6OSO3)GlcNAcβ-Sp8 1090 771 385 71
184 GlcAβ-Sp8 1072 476 238 44
282 Galβ1-4(Fucα1-3)GlcNAcβ1-3Galβ1-3(Fucα1-4)GlcNAcβ- 1062 239 120 23
Sp0
2 Neu5Acα2-8Neu5Acβ-Sp17 1060 84 42 8
174 GlcNAcβ1-6(Galβ1-3)GalNAcα-Sp8 1039 913 456 88
261 Neu5Gcα2-3Galβ1-4Glcβ-Sp0 1034 440 220 43
18 β-D-Glc-Sp8 1024 335 167 33
217 Neu5Acα2-3Galβ1-3(Fucα1-4)GlcNAcβ-Sp8 1023 646 323 63
260 Neu5Gcα2-3Galβ1-4GlcNAcβ-Sp0 1020 208 104 20
104 Galα1-3Galβ1-3GlcNAcβ-Sp0 1017 297 149 29
245 Neu5Acα2-6Galβ1-4GlcNAcβ-Sp0 1010 394 197 39
14 α-Neu5Ac-Sp8 998 1046 523 105
283 Galβ1-4GlcNAcβ1-3Galβ1-3GlcNAcβ-Sp0 978 514 257 53
156 GlcNAcα1-3Galβ1-4GlcNAcβ-Sp8 969 276 138 29
310 Manα1-3(Manα1-6)Manβ1-4GlcNAcβ1-4GlcNAcβ-Sp12 965 238 119 25
183 GlcAa-Sp8 960 463 232 48
138 Galβ1-4(Fucα1-3)GlcNAcβ1-4Galβ1-4(Fucα1-3)GlcNAcβ1- 948 595 297 63
4Galβ1-4(Fucα1-3)GlcNAcβ-Sp0
96 Galα1-3(Fucα1-2)Galβ1-4(Fucα1-3)GlcNAcβ-Sp0 948 260 130 27
6 Neu5Acα2-6Galβ1-4GlcNAcβ1-2Manα1-3(Neu5Acα2- 943 351 176 37
6Galβ1-4GlcNAcβ1-2Manα1-6)Manβ1-4GlcNAcβ1-
4GlcNAcβ-Sp12
306 GlcNAcβ1-3Man-Sp10 938 153 77 16
121 Galβ1-3(GlcNAcβ1-6)GalNAcα-Sp8 936 748 374 80
258 Neu5Gcα2-3Galβ1-3GlcNAcβ-Sp0 932 375 188 40
246 Neu5Acα2-6Galβ1-4GlcNAcβ-Sp8 931 635 317 68
200 Manβ1-4GlcNAcβ-Sp0 920 322 161 35
78 Fucβ1-3GlcNAcβ-Sp8 911 464 232 51
94 Galα1-2Galβ-Sp8 911 393 197 43
256 Galβ1-4GlcNAcβ1-2Manα1-3(Neu5Acα2-6Galβ1- 909 428 214 47
4GlcNAcβ1-2Manα1-6)Manβ1-4GlcNAcβ1-4GlcNAcβ-Sp21
95 Galα1-3(Fucα1-2)Galβ1-3GlcNAcβ-Sp0 908 245 123 27
8 α-D-Glc-Sp8 904 417 209 46
103 Galα1-3Galβ1-4(Fucα1-3)GlcNAcβ-Sp8 893 445 222 50
118 Galβ1-3(Fucα1-4)GlcNAc-Sp8 890 624 312 70
9 α-D-Man-Sp8 881 403 201 46
16 β-Neu5Ac-Sp8 876 935 468 107
119 Galβ1-3(Fucα1-4)GlcNAcβ-Sp8 872 283 141 32
278 Galβ1-3GalNAc-Sp14 851 144 72 17
187 KDNα2-3Galβ1-3GlcNAcβ-Sp0 839 386 193 46
69 Fucα1-2Galβ1-4GlcNAcβ1-3Galβ1-4GlcNAc-Sp0 837 328 164 39
76 Fucα1-3GlcNAcβ-Sp8 836 276 138 33
108 Galα1-4(Fucα1-2)Galβ1-4GlcNAcβ-Sp8 819 58 29 7
212 NeuAcα2-3(NeuAcα2-3Galβ1-3GalNAcβ1-4)Galβ1-4Glcβ- 818 1442 721 176
Sp0
132 Galβ1-3GlcNAcβ1-3Galβ1-4Glcβ-Sp10 816 353 176 43
105 Galα1-3Galβ1-4GlcNAcβ-Sp8 806 184 92 23
308 GlcNAcβ1-4GlcNAcβ-Sp12 796 360 180 45
160 GlcNAcβ1-3(GlcNAcβ1-6)Galβ1-4GlcNAcβ-Sp8 794 416 208 52
284 Neu5Acα2-3Galβ1-3GlcNAcβ1-3Galβ1-3GlcNAcβ-Sp0 777 491 245 63
188 KDNα2-3Galβ1-4GlcNAcβ-Sp0 774 320 160 41
215 Neu5Acα2-3GalNAcβ1-4GlcNAcβ-Sp0 762 252 126 33
294 Galβ1-3Galβ1-4GlcNAcβ-Sp8 746 255 128 34
196 Manα1-3(Manα1-2Manα1-2Manα1-6)Manα-Sp9 744 177 88 24
189 Manα1-2Manα1-2Manα1-3Mana-Sp9 743 207 103 28
25 GlcNAcβ1-3(GlcNAcβ1-4)(GlcNAcβ1-6)GlcNAc-Sp8 735 270 135 37
131 Galβ1-3GlcNAcβ1-3Galβ1-4GlcNAcβ-Sp0 728 290 145 40
277 Galβ1-3-(Neu5Aa2-3Galβ1-4GlcNacβ1-6)GalNAc-Sp14 722 324 162 45
136 Galβ1-4(Fucα1-3)GlcNAcβ-Sp8 718 93 46 13
70 Fucα1-2Galβ1-4GlcNAcβ1-3Galβ1-4GlcNAcβ1-3Galβ1- 713 861 430 121
4GlcNAcβ-Sp0
110 Galα1-4Galβ1-4GlcNAcβ-Sp8 712 183 91 26
129 Galβ1-3GalNAcβ1-4Galβ1-4Glcβ-Sp8 702 224 112 32
71 Fucα1-2Galβ1-4GlcNAcβ-Sp0 686 160 80 23
169 GlcNAcβ1-4(GlcNAcβ1-6)GalNAcα-Sp8 686 229 115 33
122 Galβ1-3(Neu5Acα2-6)GalNAcα-Sp8 679 157 79 23
106 Galα1-3Galβ1-4Glcβ-Sp0 678 137 69 20
255 Neu5Acβ2-6Galβ1-4GlcNAcβ-Sp8 671 153 76 23
130 Galβ1-3Galβ-Sp8 668 285 143 43
144 Galβ1-4GlcNAcβ1-3GalNAcα-Sp8 663 227 113 34
13 α-L-Rhα-Sp8 662 245 123 37
22 β-GlcNAc-Sp8 655 313 157 48
72 Fucα1-2Galβ1-4GlcNAcβ-Sp8 646 95 47 15
157 GlcNAcα1-6Galβ1-4GlcNAcβ-Sp8 644 323 162 50
307 GlcNAcβ1-4GlcNAcβ-Sp10 640 336 168 53
180 Glcβ1-4Glcβ-Sp8 608 316 158 52
191 Manα1-2Manα1-3Manα-Sp9 607 104 52 17
134 Galβ1-3GlcNAcβ-Sp8 603 103 51 17
21 β-GlcNAc-Sp0 595 285 142 48
24 (Galβ1-4GlcNAcβ)2-3,6-GalNAcα-Sp8 590 240 120 41
223 NeuAcα2-3Galβ1-3GalNAcβ1-3Galα1-4Galβ1-4Glcβ-Sp0 580 191 95 33
162 GlcNAcβ1-3Galβ-Sp8 577 435 217 75
135 Galβ1-4(Fucα1-3)GlcNAcβ-Sp0 561 139 70 25
249 Neu5Acα2-6Galβ1-4Glcβ-Sp0 560 377 189 67
48 9NAcNeu5Acα-Sp8 556 470 235 85
158 GlcNAcβ1-2Galβ1-3GalNAcα-Sp8 550 417 208 76
264 Neu5Gcα-Sp8 550 305 152 55
46 NeuAcα2-3[6OSO3]Galβ1-4GlcNAcβ-Sp8 545 363 182 67
68 Fucα1-2Galβ1-4(Fucα1-3)GlcNAcβ-Sp8 541 208 104 38
222 Neu5Acα2-3Galβ-Sp8 526 277 139 53
298 Galβ1-4GlcNAcα1-6Galβ1-4GlcNAcβ-Sp0 494 335 167 68
98 Galα1-3(Fucα1-2)Galβ1-4Glcβ-Sp0 482 112 56 23
312 Manα1-6(Manα1-3)Manα1-6(Manα1-3)Manβ-Sp10 453 292 146 64
133 Galβ1-3GlcNAcβ-Sp0 452 165 82 36
57 Fucα1-2Galβ1-3(Fucα1-4)GlcNAcβ-Sp8 450 268 134 60
114 Galβ1-2Galβ-Sp8 449 324 162 72
198 Manα1-6(Manα1-3)Manα1-6(Manα1-3)Manβ1-4GlcNAcβ1-4 448 204 102 45
GlcNAcβ-Sp12
161 GlcNAcβ1-3GalNAcα-Sp8 442 156 78 35
281 Galβ1-4[Fucα1-3][6OSO3]Glc-Sp0 439 144 72 33
259 Neu5Gcα2-3Galβ1-4(Fucα1-3)GlcNAcβ-Sp0 433 357 179 83
67 Fucα1-2Galβ1-4(Fucα1-3)GlcNAcβ-Sp0 420 94 47 22
12 α-L-Fuc-Sp9 410 303 151 74
159 GlcNAcβ1-3(GlcNAcβ1-6)GalNAcα-Sp8 407 88 44 22
75 Fucα1-3GlcNAcβ-Sp8 399 182 91 46
239 Neu5Acα2-3Galβ1-4Glcβ-Sp0 395 156 78 39
290 Galα1-3(Fucα1-2)Galβ-Sp18 389 246 123 63
11 α-L-Fuc-Sp8 387 231 115 60
51 GlcNAcβ1-2Manα1-3(GlcNAcβ1-2Manα1-6)Manβ1- 383 164 82 43
4GlcNAcβ1-4GlcNAcβ-Sp13
5 Galβ1-3GlcNAcβ1-2Manα1-3(Galβ1-3GlcNAcβ1-2Manα1-6) 381 529 265 139
Manβ1-4GlcNAcβ1-4GlcNAcβ-Sp19
63 Fucα1-2Galβ1-3GlcNAcβ-Sp0 362 187 93 52
241 Galβ1-4GlcNAcβ1-2Manα1-3(Fucα1-3(Galβ1-4)GlcNAcβ1- 352 68 34 19
2Manα1-6)Manβ1-4GlcNAcβ1-4GlcNAcβ-Sp20
155 Galβ1-4Glcβ-Sp8 315 105 53 33
126 Galβ1-3GalNAcβ-Sp8 288 265 132 92
195 Manα1-3(Manα1-6)Manα-Sp9 269 92 46 34
88 GalNAcβ1-3GalNAcα-Sp8 262 107 54 41
252 Neu5Acα2-8Neu5Acα-Sp8 260 214 107 82
167 GlcNAcβ1-3Galβ1-4Glcβ-Sp0 257 129 64 50
140 Galβ1-4[6OSO3]Glcβ-Sp8 256 345 172 135
177 Glcα1-4Glcβ-Sp8 246 113 57 46
179 Glcα1-6Glcα1-6Glcβ-Sp8 225 380 190 168
314 Manα1-2Manα1-2Manα1-3(Manα1-2Manα1-6(Manα1- 221 329 165 149
2Manα1-3)Manα1-6)Manα-Sp9
238 Neu5Acα2-3Galβ1-4GlcNAcβ1-3Galβ1-4GlcNAcβ-Sp0 212 200 100 94
220 Neu5Acα2-3Galβ1-3[6OSO3]GalNAcα-Sp8 210 153 77 73
142 Galβ1-4GalNAcβ1-3(Fucα1-2)Galβ1-4GlcNAcβ-Sp8 204 126 63 62
61 Fucα1-2Galβ1-3GlcNAcβ1-3Galβ1-4Glcβ-Sp10 196 67 34 34
102 Galα1-3GalNAcβ-Sp8 188 198 99 105
170 GlcNAcβ1-4Galβ1-4GlcNAcβ-Sp8 184 127 64 69
124 Galβ1-3(Neu5Acα2-6)GlcNAcβ1-4Galβ1-4Glcβ-Sp10 173 146 73 84
100 Galα1-3(Galα1-4)Galβ1-4GlcNAcβ-Sp8 168 112 56 66
186 GlcAβ1-6Galβ-Sp8 158 171 86 108
4 Neu5Gcβ2-6Galβ1-4GlcNAc-Sp8 152 96 48 63
73 Fucα1-2Galβ1-4Glcβ-Sp0 148 205 103 139
49 9NAcNeu5Acα2-6Galβ1-4GlcNAcβ-Sp8 146 159 79 108
58 Fucα1-2Galβ1-3GalNAcα-Sp8 136 171 86 126
250 Neu5Acα2-6Galβ1-4Glcβ-Sp8 122 144 72 119
112 Galα1-4GlcNAcβ-Sp8 115 82 41 72
165 GlcNAcβ1-3Galβ1-4GlcNAcβ-Sp8 84 68 34 81
226 Neu5Acα2-3Galβ1-3GlcNAcβ-Sp8 76 85 42 112
288 [6OSO3]Galβ1-4[6OSO3]GlcNacβ-Sp0 72 130 65 180
153 Galβ1-4GlcNAcβ-Sp8 48 58 29 120
It is contemplated that any embodiment discussed in this specification can be implemented with respect to any method, kit, reagent, or composition of the invention, and vice versa. Furthermore, compositions of the invention can be used to achieve methods of the invention.
It will be understood that particular embodiments described herein are shown by way of illustration and not as limitations of the invention. The principal features of this invention can be employed in various embodiments without departing from the scope of the invention. Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, numerous equivalents to the specific procedures described herein. Such equivalents are considered to be within the scope of this invention and are covered by the claims.
All publications and patent applications mentioned in the specification are indicative of the level of skill of those skilled in the art to which this invention pertains. All publications and patent applications are herein incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference.
The use of the word “a” or “an” when used in conjunction with the term “comprising” in the claims and/or the specification may mean “one,” but it is also consistent with the meaning of “one or more,” “at least one,” and “one or more than one.” The use of the term “or” in the claims is used to mean “and/or” unless explicitly indicated to refer to alternatives only or the alternatives are mutually exclusive, although the disclosure supports a definition that refers to only alternatives and “and/or.” Throughout this application, the term “about” is used to indicate that a value includes the inherent variation of error for the device, the method being employed to determine the value, or the variation that exists among the study subjects.
As used in this specification and claim(s), the words “comprising” (and any form of comprising, such as “comprise” and “comprises”), “having” (and any form of having, such as “have” and “has”), “including” (and any form of including, such as “includes” and “include”) or “containing” (and any form of containing, such as “contains” and “contain”) are inclusive or open-ended and do not exclude additional, unrecited elements or method steps.
The term “or combinations thereof” as used herein refers to all permutations and combinations of the listed items preceding the term. For example, “A, B, C, or combinations thereof” is intended to include at least one of: A, B, C, AB, AC, BC, or ABC, and if order is important in a particular context, also BA, CA, CB, CBA, BCA, ACB, BAC, or CAB. Continuing with this example, expressly included are combinations that contain repeats of one or more item or term, such as BB, AAA, MB, BBC, AAABCCCC, CBBAAA, CABABB, and so forth. The skilled artisan will understand that typically there is no limit on the number of items or terms in any combination, unless otherwise apparent from the context.
All of the compositions and/or methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. While the compositions and methods of this invention have been described in terms of preferred embodiments, it will be apparent to those of skill in the art that variations may be applied to the compositions and/or methods and in the steps or in the sequence of steps of the method described herein without departing from the concept, spirit and scope of the invention. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention as defined by the appended claims.
REFERENCES
  • 1. C. G. Figdor, Y. van Kooyk, G. J. Adema, Nat Rev Immunol 2, 77 (February, 2002).
  • 2. E. Pyz, A. S. Marshall, S. Gordon, G. D. Brown, Ann Med 38, 242 (2006).
  • 3. G. D. Brown, Nat Rev Immunol 6, 33 (January, 2006).
  • 4. T. B. Geijtenbeek et al., Nat Immunol 1, 353 (October, 2000).
  • 5. T. B. Geijtenbeek et al., Cell 100, 575 (Mar. 3, 2000).
  • 6. C. F. d'Ostiani et al., J Exp Med 191, 1661 (May 15, 2000).
  • 7. C. Fradin, D. Poulain, T. Jouault, Infect Immun 68, 4391 (August, 2000).
  • 8. A. Cambi et al., Eur J Immunol 33, 532 (February, 2003).
  • 9. M. G. Netea, J. W. Meer, I. Verschueren, B. J. Kullberg, Eur J Immunol 32, 1455 (May, 2002).
  • 10. S. J. Lee et al., Science 295, 1898 (Mar. 8, 2002).
  • 11. N. Maeda et al., J Biol Chem 278, 5513 (Feb. 21, 2003).
  • 12. L. Tailleux et al., J Exp Med 197, 121 (Jan. 6, 2003).
  • 13. T. B. Geijtenbeek et al., J Exp Med 197, 7 (Jan. 6, 2003).
  • 14. A. M. Cooper et al., J Immunol 168, 1322 (Feb. 1, 2002).
  • 15. J. Banchereau et al., Annu Rev Immunol 18, 767 (2000).
  • 16. P. Jeannin et al., Immunity 22, 551 (May, 2005).
  • 17. B. N. Gantner, R. M. Simmons, S. J. Canavera, S. Akira, D. M. Underhill, J Exp Med 197, 1107 (May 5, 2003).
  • 18. H. Moriwaki et al., FEBS Lett 440, 29 (Nov. 27, 1998).
  • 19. M. Wykes, G. MacPherson, Immunology 100, 1 (May, 2000).
  • 20. M. Balazs, F. Martin, T. Zhou, J. Kearney, Immunity 17, 341 (September, 2002).
  • 21. T. Kikuchi, S. Worgall, R. Singh, M. A. Moore, R. G. Crystal, Nat Med 6, 1154 (October, 2000).
  • 22. B. Dubois et al., J Leukoc Biol 66, 224 (August, 1999).
  • 23. H. Qi, J. G. Egen, A. Y. Huang, R. N. Germain, Science 312, 1672 (Jun. 16, 2006).
  • 24. A. Bergtold, D. D. Desai, A. Gavhane, R. Clynes, Immunity 23, 503 (November, 2005).
  • 25. C. R. Ruprecht, A. Lanzavecchia, Eur J Immunol 36, 810 (April, 2006).
  • 26. N. L. Bernasconi, E. Traggiai, A. Lanzavecchia, Science 298, 2199 (Dec. 13, 2002).
  • 27. P. A. Moore et al., Science 285, 260 (Jul. 9, 1999).
  • 28. J. A. Gross et al., Nature 404, 995 (Apr. 27, 2000).
  • 29. A. Craxton, D. Magaletti, E. J. Ryan, E. A. Clark, Blood 101, 4464 (Jun. 1, 2003).
  • 30. I. MacLennan, C. Vinuesa, Immunity 17, 235 (September, 2002).
  • 31. J. Banchereau, R. M. Steinman, Nature 392, 245 (Mar. 19, 1998).
  • 32. J. Banchereau, P. de Paoli, A. Valle, E. Garcia, F. Rousset, Science 251, 70 (Jan. 4, 1991).
  • 33. B. Beutler et al., Annu Rev Immunol 24, 353 (2006).
  • 34. K. Hayashida, N. Kume, M. Minami, T. Kita, FEBS Lett 511, 133 (Jan. 30, 2002).
  • 35. M. Kakutani, T. Masaki, T. Sawamura, Proc Natl Acad Sci USA 97, 360 (Jan. 4, 2000).
  • 36. M. Colonna, J. Samaridis, L. Angman, Eur J Immunol 30, 697 (February, 2000).
  • 37. J. Valladeau et al., J Immunol 167, 5767 (Nov. 15, 2001).
  • 38. G. Jego et al., Immunity 19, 225 (August, 2003).
  • 39. M. N. Wykes, L. Beattie, G. G. Macpherson, D. N. Hart, Immunology 113, 318 (November, 2004).
  • 40. Y. Delneste et al., Immunity 17, 353 (September, 2002).
  • 41. Y. Wang et al., Immunity 15, 971 (December, 2001).
  • 42. A. Asea et al., Nat Med 6, 435 (April, 2000).
  • 43. H. Bausinger et al., Eur J Immunol 32, 3708 (December, 2002).
  • 44. E. E. Bates et al., J Immunol 163, 1973 (Aug. 15, 1999).

Claims (10)

What is claimed is:
1. A method of decreasing an antibody-mediated immune response to an autoantigen in a subject, comprising the step of administering to the subject an effective amount of a composition comprising a recombinant fusion protein, said fusion protein comprising an anti-human dendritic cell asialoglycoprotein receptor (ASGPR) antibody or an antigen binding fragment thereof and one or more autoimmune antigens, wherein the antibody or fragment thereof comprises (a) an immunoglobulin heavy chain selected from the group consisting of SEQ ID NO: 5, 7, 9, and 11; and (b) an immunoglobulin light chain selected from the group consisting of SEQ ID NO: 6, 8, 10, and 12.
2. The method of claim 1, wherein the subject has an autoimmune disorder.
3. The method of claim 2, wherein the autoimmune disorder is multiple sclerosis.
4. The method of claim 2, wherein the autoimmune disorder is systemic lupus erythematosus (SLE).
5. The method of claim 2, wherein the autoimmune disorder is myasthenia gravis (MG).
6. The method of claim 1, wherein the autoantigen is Smith protein, RNP ribonucleoprotein, SS-A protein, SS-B protein, acetylcholine receptor, luteinizing hormone, follicular stimulating hormone, testosterone, growth hormone, prolactin, glutamic acid decarboxylase 65 (GAD 65), myelin basic protein, myelin proteolipid protein, thyroglobulin, or thyroid stimulating hormone (TSH) receptor.
7. The method of claim 1, wherein the antibody or fragment thereof comprises SEQ ID NO: 5 and SEQ ID NO: 6.
8. The method of claim 1, wherein the antibody or fragment thereof comprises SEQ ID NO: 7 and SEQ ID NO: 8.
9. The method of claim 1, wherein the antibody or fragment thereof comprises SEQ ID NO: 9 and SEQ ID NO: 10.
10. The method of claim 1, wherein the antibody or fragment thereof comprises SEQ ID NO: 11 and SEQ ID NO: 12.
US16/372,832 2007-02-02 2019-04-02 Agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (DC-ASGPR) Active 2028-05-07 US11173202B2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US16/372,832 US11173202B2 (en) 2007-02-02 2019-04-02 Agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (DC-ASGPR)
US17/454,108 US20220152192A1 (en) 2007-02-02 2021-11-09 Agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (dc-asgpr)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US88803607P 2007-02-02 2007-02-02
US12/025,010 US8236934B2 (en) 2007-02-02 2008-02-02 Agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (DC-ASGPR)
US13/551,198 US8728481B2 (en) 2007-02-02 2012-07-17 Agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (DC-ASGPR)
US14/254,206 US9453074B2 (en) 2007-02-02 2014-04-16 Agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (DC-ASGPR)
US15/265,236 US10279030B2 (en) 2007-02-02 2016-09-14 Agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (DC-ASGPR)
US16/372,832 US11173202B2 (en) 2007-02-02 2019-04-02 Agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (DC-ASGPR)

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US15/265,236 Continuation US10279030B2 (en) 2007-02-02 2016-09-14 Agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (DC-ASGPR)

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/454,108 Continuation US20220152192A1 (en) 2007-02-02 2021-11-09 Agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (dc-asgpr)

Publications (2)

Publication Number Publication Date
US20190231865A1 US20190231865A1 (en) 2019-08-01
US11173202B2 true US11173202B2 (en) 2021-11-16

Family

ID=39682353

Family Applications (6)

Application Number Title Priority Date Filing Date
US12/025,010 Active 2028-07-13 US8236934B2 (en) 2007-02-02 2008-02-02 Agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (DC-ASGPR)
US13/551,198 Active US8728481B2 (en) 2007-02-02 2012-07-17 Agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (DC-ASGPR)
US14/254,206 Active 2028-02-28 US9453074B2 (en) 2007-02-02 2014-04-16 Agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (DC-ASGPR)
US15/265,236 Active 2028-03-29 US10279030B2 (en) 2007-02-02 2016-09-14 Agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (DC-ASGPR)
US16/372,832 Active 2028-05-07 US11173202B2 (en) 2007-02-02 2019-04-02 Agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (DC-ASGPR)
US17/454,108 Pending US20220152192A1 (en) 2007-02-02 2021-11-09 Agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (dc-asgpr)

Family Applications Before (4)

Application Number Title Priority Date Filing Date
US12/025,010 Active 2028-07-13 US8236934B2 (en) 2007-02-02 2008-02-02 Agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (DC-ASGPR)
US13/551,198 Active US8728481B2 (en) 2007-02-02 2012-07-17 Agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (DC-ASGPR)
US14/254,206 Active 2028-02-28 US9453074B2 (en) 2007-02-02 2014-04-16 Agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (DC-ASGPR)
US15/265,236 Active 2028-03-29 US10279030B2 (en) 2007-02-02 2016-09-14 Agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (DC-ASGPR)

Family Applications After (1)

Application Number Title Priority Date Filing Date
US17/454,108 Pending US20220152192A1 (en) 2007-02-02 2021-11-09 Agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (dc-asgpr)

Country Status (14)

Country Link
US (6) US8236934B2 (en)
EP (1) EP2115129B1 (en)
JP (1) JP2010518024A (en)
KR (1) KR20090114432A (en)
CN (1) CN101952414A (en)
AU (1) AU2008214036B2 (en)
BR (1) BRPI0807152A2 (en)
CA (1) CA2715045C (en)
IL (2) IL200093A0 (en)
MX (1) MX2009008211A (en)
NZ (2) NZ593639A (en)
TW (1) TWI422594B (en)
WO (1) WO2008097870A2 (en)
ZA (1) ZA200905274B (en)

Families Citing this family (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI422594B (en) 2007-02-02 2014-01-11 Baylor Res Inst Agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (dc-asgpr)
BRPI0807344A2 (en) * 2007-02-02 2014-05-20 Baylor Res Inst MULTI-VARIABLE ANTIGENS COMPLEXED WITH HUMANIZED MONOCLONAL STEERING ANTIBODY
EP3219732A1 (en) * 2009-03-10 2017-09-20 Baylor Research Institute Antigen presenting cell targeted cancer vaccines
CA2754764A1 (en) 2009-03-10 2010-09-16 Baylor Research Institute Antigen presenting cell targeted cancer vaccines
US9562104B2 (en) 2009-03-10 2017-02-07 Baylor Research Institute Anti-CD40 antibodies
RU2012152828A (en) * 2010-05-07 2014-06-20 Бейлор Рисёч Инститьют Mediated by Dendritic Cell Immunoreceptors (DCIR) Cross-Priming of Human CD8 + T Cells
US9850296B2 (en) 2010-08-10 2017-12-26 Ecole Polytechnique Federale De Lausanne (Epfl) Erythrocyte-binding therapeutics
US9517257B2 (en) 2010-08-10 2016-12-13 Ecole Polytechnique Federale De Lausanne (Epfl) Erythrocyte-binding therapeutics
US9518087B2 (en) 2010-08-10 2016-12-13 Ecole Polytechnique Federale De Lausanne (Epfl) Erythrocyte-binding therapeutics
US20120121592A1 (en) * 2010-10-13 2012-05-17 Baylor Research Institute Targeting Antigens to Human Dendritic Cells Via DC-Asialoglycoprotein Receptor to Produce IL-10 Regulatory T-Cells
WO2012129227A1 (en) * 2011-03-22 2012-09-27 Baylor Research Institute Dendritic cells (dcs) targeting for tuberculosis (tb) vaccine
AR085573A1 (en) * 2011-03-25 2013-10-09 Baylor Res Inst COMPOSITIONS AND IMMUNIZATION METHODS AGAINST THE VIRUS OF HEPATITIS C
US20130017151A1 (en) * 2011-07-11 2013-01-17 Baylor Research Institute SUBSETS OF ANTIGEN-PRESENTING CELLS (APCs) IN THE HUMAN VAGINA AND THEIR DISTINCT FUNCTIONS
CN112587658A (en) 2012-07-18 2021-04-02 博笛生物科技有限公司 Targeted immunotherapy for cancer
WO2014031984A1 (en) * 2012-08-24 2014-02-27 Baylor Research Institute Immunological detection methods and compositions
WO2014085580A1 (en) * 2012-11-28 2014-06-05 Baylor Research Institute Methods and compositions involving a flu vaccine
US20160296609A1 (en) * 2013-06-28 2016-10-13 Baylor Research Institute Dendritic cell asgpr targeting immunotherapeutics for multiple sclerosis
CN103409451A (en) * 2013-06-28 2013-11-27 扬州维克斯生物科技有限公司 Method for loading tumor antigen peptide to dendritic cell (DC) in targeting manner
MX2016008520A (en) * 2013-12-24 2016-11-30 Astellas Pharma Inc Novel anti-human bdca-2 antibody.
AU2015205753A1 (en) 2014-01-10 2016-07-21 Birdie Biopharmaceuticals Inc. Compounds and compositions for treating HER2 positive tumors
CN106103483A (en) 2014-01-13 2016-11-09 贝勒研究院 The novel vaccine of the related disease of anti-HPV with HPV
US10046056B2 (en) 2014-02-21 2018-08-14 École Polytechnique Fédérale De Lausanne (Epfl) Glycotargeting therapeutics
SG11201606761RA (en) 2014-02-21 2016-09-29 Ecole Polytecnique Federale De Lausanne Epfl Epfl Tto Glycotargeting therapeutics
US10946079B2 (en) 2014-02-21 2021-03-16 Ecole Polytechnique Federale De Lausanne Glycotargeting therapeutics
US10953101B2 (en) 2014-02-21 2021-03-23 École Polytechnique Fédérale De Lausanne (Epfl) Glycotargeting therapeutics
CN110845615B (en) 2014-03-21 2023-10-20 艾伯维公司 Anti-EGFR antibodies and antibody drug conjugates
US10993990B2 (en) * 2014-05-16 2021-05-04 Baylor Research Institute Methods and compositions for treating autoimmune and inflammatory conditions
DK3166976T3 (en) 2014-07-09 2022-04-11 Birdie Biopharmaceuticals Inc ANTI-PD-L1 COMBINATIONS FOR TREATMENT OF TUMORS
CN112546238A (en) 2014-09-01 2021-03-26 博笛生物科技有限公司 anti-PD-L1 conjugates for the treatment of tumors
WO2016118787A1 (en) * 2015-01-21 2016-07-28 University Of Florida Research Foundation, Inc. Engineered receptor/ligand system for delivery of therapeutic agents
CN106943598A (en) 2016-01-07 2017-07-14 博笛生物科技(北京)有限公司 Anti- HER2 for treating tumour is combined
CN115554406A (en) 2016-01-07 2023-01-03 博笛生物科技有限公司 anti-CD 20 combinations for the treatment of tumors
CN115252792A (en) 2016-01-07 2022-11-01 博笛生物科技有限公司 anti-EGFR combinations for the treatment of tumors
MX2018015274A (en) 2016-06-08 2019-10-07 Abbvie Inc Anti-cd98 antibodies and antibody drug conjugates.
US20200002421A1 (en) 2016-06-08 2020-01-02 Abbvie Inc. Anti-b7-h3 antibodies and antibody drug conjugates
EP3469000A1 (en) 2016-06-08 2019-04-17 AbbVie Inc. Anti-b7-h3 antibodies and antibody drug conjugates
TW202304996A (en) 2016-06-08 2023-02-01 美商艾伯維有限公司 Anti-b7-h3 antibodies and antibody drug conjugates
JP7237843B2 (en) 2017-02-21 2023-03-13 ユニバーシティー オブ フロリダ リサーチ ファンデーション, インク. Modified AAV capsid proteins and uses thereof
EP3612567A4 (en) 2017-04-19 2020-11-11 Bluefin Biomedicine, Inc. Anti-vtcn1 antibodies and antibody drug conjugates
CN108794467A (en) 2017-04-27 2018-11-13 博笛生物科技有限公司 2- amino-quinoline derivatives
US11253579B2 (en) 2017-06-16 2022-02-22 The University Of Chicago Compositions and methods for inducing immune tolerance
EP3641771A4 (en) 2017-06-23 2020-12-16 Birdie Biopharmaceuticals, Inc. Pharmaceutical compositions
US10610585B2 (en) 2017-09-26 2020-04-07 Inserm (Institut National De La Sante Et De La Recherche Medicale) Methods and compositions for treating and preventing HIV
AR124681A1 (en) 2021-01-20 2023-04-26 Abbvie Inc ANTI-EGFR ANTIBODY-DRUG CONJUGATES

Citations (46)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3791932A (en) 1971-02-10 1974-02-12 Akzona Inc Process for the demonstration and determination of reaction components having specific binding affinity for each other
US3949064A (en) 1973-10-26 1976-04-06 Baxter Laboratories, Inc. Method of detecting antigens or antibodies
US4174384A (en) 1975-06-30 1979-11-13 Syva Company Fluorescence quenching with immunological pairs in immunoassays
US4554101A (en) 1981-01-09 1985-11-19 New York Blood Center, Inc. Identification and preparation of epitopes on antigens and allergens on the basis of hydrophilicity
WO1997014789A2 (en) 1995-10-17 1997-04-24 Genencor International, Inc. Enzymatic array and process of making same
FR2748479A1 (en) 1996-05-10 1997-11-14 Pasteur Institut Cellulase proteins with cohesin or dockerin type II domains
US5738985A (en) 1993-04-02 1998-04-14 Ribogene, Inc. Method for selective inactivation of viral replication
US5945308A (en) 1998-04-03 1999-08-31 Incyte Pharmaceuticals, Inc. Human oxidized LDL receptor
US6046158A (en) 1996-12-20 2000-04-04 Board Of Regents The University Of Texas Systems Unique dendritic cell-associated C-type lectins, dectin-1 and dectin-2; compositions and uses thereof
JP2000157282A (en) 1998-11-30 2000-06-13 Toyota Central Res & Dev Lab Inc Enzyme array complex and immobilized enzyme array complex and production of these, carrier molecule, and enzyme
WO2000063151A1 (en) 1999-04-21 2000-10-26 Basf Aktiengesellschaft Mixture of adipic or phthalic acid diesters and isomeric nonanols
WO2000063251A1 (en) 1999-04-19 2000-10-26 Katholieke Universiteit Nijmegen Composition and method for modulating dendritic cell-t cell interaction
US6277959B1 (en) 1997-07-09 2001-08-21 Schering Corporation Isolated mammalian membrane protein genes; related reagents
WO2002035242A1 (en) 2000-10-27 2002-05-02 Pierre Fabre Medicament Method for identifying novel molecules binding with lox receptor
US20020055618A1 (en) 1997-10-07 2002-05-09 William H.R. Langridge Methods and substances for preventing and treating autoimmune disease
US6410241B1 (en) 1999-03-24 2002-06-25 Board Of Regents, The University Of Texas System Methods of screening open reading frames to determine whether they encode polypeptides with an ability to generate an immune response
US6451995B1 (en) 1996-03-20 2002-09-17 Sloan-Kettering Institute For Cancer Research Single chain FV polynucleotide or peptide constructs of anti-ganglioside GD2 antibodies, cells expressing same and related methods
US6541011B2 (en) 1998-02-11 2003-04-01 Maxygen, Inc. Antigen library immunization
WO2003036895A1 (en) 2001-10-25 2003-05-01 Renesas Technology Corp. Semiconductor integrated circuit for communication, modulating/demodulating device, and communication diagnosing method
WO2003073827A2 (en) 2002-02-28 2003-09-12 Corixa Corporation Methods of modulating dendritic cells using adjuvants
EP1418234A1 (en) 2000-03-02 2004-05-12 Abgenix, Inc. Human monoclonal antibodies against oxidized ldl receptor and medicinal use thereof
US6738985B2 (en) 2002-05-14 2004-05-25 David S. Hahn Disposable sweatband liner
US20040126357A1 (en) 2002-08-20 2004-07-01 Genitrix, Llc Lectin compositions and methods for modulating an immune response to an antigen
EP1441030A1 (en) 2001-10-11 2004-07-28 Katakura Industries Co., Ltd. Method of purifying recombinant fused protein and method of producing protein using the same
JP2004236504A (en) 2003-02-03 2004-08-26 Toyota Central Res & Dev Lab Inc Enzyme array complex and immobilized enzyme array complex, method for producing the same, carrier molecule and enzyme
US20040258688A1 (en) 1995-01-31 2004-12-23 Daniel Hawiger Enhanced antigen delivery and modulation of the immune response therefrom
US20040265901A1 (en) 2001-08-22 2004-12-30 Shengfeng Li Compositions and methods for generating antigen-binding units
US20050037001A1 (en) 2001-11-30 2005-02-17 Germeraad Wilfred Thomas Vincent APC targeting conjugate, an antigen-presenting cell contacted with such conjugate, their medical use, and methods of production
US20050064509A1 (en) 2003-09-23 2005-03-24 The Regents Of The University Of California Use of templated self assembly to create novel multifunctional species
WO2006004663A2 (en) 2004-06-25 2006-01-12 Medimmune, Inc. Increasing the production of recombinant antibodies in mammalian cells by site-directed mutagenesis
WO2006107617A2 (en) 2005-04-06 2006-10-12 Ibc Pharmaceuticals, Inc. Methods for generating stably linked complexes composed of homodimers, homotetramers or dimers of dimers and uses
US20060257412A1 (en) 2003-03-04 2006-11-16 Bowdish Katherine S Method of treating autoimmune disease by inducing antigen presentation by tolerance inducing antigen presenting cells
US20060269949A1 (en) 2005-05-23 2006-11-30 Halloran Philip F Tissue rejection
WO2008097817A2 (en) 2007-02-02 2008-08-14 Baylor Research Institute Multivariable antigens complexed with targeting humanized monoclonal antibody
US20080233140A1 (en) 2007-02-23 2008-09-25 Baylor Research Institute Therapeutic Applications of Activation of Human Antigen-Presenting Cells Through Dectin-1
US20090186025A1 (en) 2006-03-22 2009-07-23 Immunobiology Limited Fusion Protein Comprising an Fc Receptor Binding Polypeptide and an Antigenic Polypeptide for Mediating an Immune Response
US7592003B2 (en) 2005-09-30 2009-09-22 Oklahoma Medical Research Foundation Regulation of toll-like receptors on stem cells
US20120039916A1 (en) 2010-08-13 2012-02-16 Baylor Research Institute Novel vaccine adjuvants based on targeting adjuvants to antibodies directly to antigen-presenting cells
US20120121592A1 (en) 2010-10-13 2012-05-17 Baylor Research Institute Targeting Antigens to Human Dendritic Cells Via DC-Asialoglycoprotein Receptor to Produce IL-10 Regulatory T-Cells
US8236934B2 (en) 2007-02-02 2012-08-07 Baylore Research Institute Agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (DC-ASGPR)
US20120231023A1 (en) 2011-03-08 2012-09-13 Baylor Research Institute Novel Vaccine Adjuvants Based on Targeting Adjuvants to Antibodies Directly to Antigen-Presenting Cells
US20120237513A1 (en) 2007-02-02 2012-09-20 Baylor Research Institute Vaccines Based on Targeting Antigen to DCIR Expressed on Antigen-Presenting Cells
US20120244155A1 (en) 2011-03-22 2012-09-27 Baylor Research Institute Dendritic Cells (DCs) Targeting for Tuberculosis (TB) Vaccine
US20120301465A1 (en) 2011-03-25 2012-11-29 Institut National De La Sante Et De La Recherche Medicale (Inserm) Compositions and methods to immunize against hepatitis c virus
US20120315269A1 (en) 2011-05-05 2012-12-13 Baylor Research Institute Immunoglobulin-like transcript (ilt) receptors as cd8 antagonists
US8481314B2 (en) 2007-02-23 2013-07-09 Baylor Research Institute Activation of human antigen-presenting cells through dendritic cell lectin-like oxidized LDL receptor-1 (LOX-1)

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7217420B2 (en) * 2002-07-13 2007-05-15 The University Of Georgia Research Foundation, Inc. Mycoplasma gallisepticum formulation

Patent Citations (59)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3791932A (en) 1971-02-10 1974-02-12 Akzona Inc Process for the demonstration and determination of reaction components having specific binding affinity for each other
US3949064A (en) 1973-10-26 1976-04-06 Baxter Laboratories, Inc. Method of detecting antigens or antibodies
US4174384A (en) 1975-06-30 1979-11-13 Syva Company Fluorescence quenching with immunological pairs in immunoassays
US4554101A (en) 1981-01-09 1985-11-19 New York Blood Center, Inc. Identification and preparation of epitopes on antigens and allergens on the basis of hydrophilicity
US5738985A (en) 1993-04-02 1998-04-14 Ribogene, Inc. Method for selective inactivation of viral replication
US20040258688A1 (en) 1995-01-31 2004-12-23 Daniel Hawiger Enhanced antigen delivery and modulation of the immune response therefrom
WO1997014789A2 (en) 1995-10-17 1997-04-24 Genencor International, Inc. Enzymatic array and process of making same
US6451995B1 (en) 1996-03-20 2002-09-17 Sloan-Kettering Institute For Cancer Research Single chain FV polynucleotide or peptide constructs of anti-ganglioside GD2 antibodies, cells expressing same and related methods
FR2748479A1 (en) 1996-05-10 1997-11-14 Pasteur Institut Cellulase proteins with cohesin or dockerin type II domains
US6046158A (en) 1996-12-20 2000-04-04 Board Of Regents The University Of Texas Systems Unique dendritic cell-associated C-type lectins, dectin-1 and dectin-2; compositions and uses thereof
US7129039B2 (en) 1996-12-20 2006-10-31 Board Of Regents The University Of Texas System Unique dendritic cell-associated C-type lectins, dectin-1 and dectin-2; compositions and uses thereof
US6277959B1 (en) 1997-07-09 2001-08-21 Schering Corporation Isolated mammalian membrane protein genes; related reagents
US20040192892A1 (en) 1997-07-09 2004-09-30 Jenny Valladeau Isolated mammalian membrane protein genes; related reagents
US20020055618A1 (en) 1997-10-07 2002-05-09 William H.R. Langridge Methods and substances for preventing and treating autoimmune disease
US6541011B2 (en) 1998-02-11 2003-04-01 Maxygen, Inc. Antigen library immunization
US5945308A (en) 1998-04-03 1999-08-31 Incyte Pharmaceuticals, Inc. Human oxidized LDL receptor
JP2000157282A (en) 1998-11-30 2000-06-13 Toyota Central Res & Dev Lab Inc Enzyme array complex and immobilized enzyme array complex and production of these, carrier molecule, and enzyme
US6410241B1 (en) 1999-03-24 2002-06-25 Board Of Regents, The University Of Texas System Methods of screening open reading frames to determine whether they encode polypeptides with an ability to generate an immune response
WO2000063251A1 (en) 1999-04-19 2000-10-26 Katholieke Universiteit Nijmegen Composition and method for modulating dendritic cell-t cell interaction
WO2000063151A1 (en) 1999-04-21 2000-10-26 Basf Aktiengesellschaft Mixture of adipic or phthalic acid diesters and isomeric nonanols
EP1418234A1 (en) 2000-03-02 2004-05-12 Abgenix, Inc. Human monoclonal antibodies against oxidized ldl receptor and medicinal use thereof
WO2002035242A1 (en) 2000-10-27 2002-05-02 Pierre Fabre Medicament Method for identifying novel molecules binding with lox receptor
US20040265901A1 (en) 2001-08-22 2004-12-30 Shengfeng Li Compositions and methods for generating antigen-binding units
US7179595B2 (en) 2001-08-22 2007-02-20 Shengfeng Li Compositions and methods for generating antigen-binding units
US20050106700A1 (en) 2001-10-11 2005-05-19 Tsuyoshi Nomura Method of purifying recombinant fused protein and method of producing protein using the same
EP1441030A1 (en) 2001-10-11 2004-07-28 Katakura Industries Co., Ltd. Method of purifying recombinant fused protein and method of producing protein using the same
WO2003036895A1 (en) 2001-10-25 2003-05-01 Renesas Technology Corp. Semiconductor integrated circuit for communication, modulating/demodulating device, and communication diagnosing method
US20050037001A1 (en) 2001-11-30 2005-02-17 Germeraad Wilfred Thomas Vincent APC targeting conjugate, an antigen-presenting cell contacted with such conjugate, their medical use, and methods of production
WO2003073827A2 (en) 2002-02-28 2003-09-12 Corixa Corporation Methods of modulating dendritic cells using adjuvants
US6738985B2 (en) 2002-05-14 2004-05-25 David S. Hahn Disposable sweatband liner
US20040126357A1 (en) 2002-08-20 2004-07-01 Genitrix, Llc Lectin compositions and methods for modulating an immune response to an antigen
JP2004236504A (en) 2003-02-03 2004-08-26 Toyota Central Res & Dev Lab Inc Enzyme array complex and immobilized enzyme array complex, method for producing the same, carrier molecule and enzyme
US20060257412A1 (en) 2003-03-04 2006-11-16 Bowdish Katherine S Method of treating autoimmune disease by inducing antigen presentation by tolerance inducing antigen presenting cells
US20050064509A1 (en) 2003-09-23 2005-03-24 The Regents Of The University Of California Use of templated self assembly to create novel multifunctional species
WO2006004663A2 (en) 2004-06-25 2006-01-12 Medimmune, Inc. Increasing the production of recombinant antibodies in mammalian cells by site-directed mutagenesis
WO2006107617A2 (en) 2005-04-06 2006-10-12 Ibc Pharmaceuticals, Inc. Methods for generating stably linked complexes composed of homodimers, homotetramers or dimers of dimers and uses
US7666596B2 (en) 2005-05-23 2010-02-23 University Of Alberta Tissue rejection
US20060269949A1 (en) 2005-05-23 2006-11-30 Halloran Philip F Tissue rejection
US7592003B2 (en) 2005-09-30 2009-09-22 Oklahoma Medical Research Foundation Regulation of toll-like receptors on stem cells
US20090186025A1 (en) 2006-03-22 2009-07-23 Immunobiology Limited Fusion Protein Comprising an Fc Receptor Binding Polypeptide and an Antigenic Polypeptide for Mediating an Immune Response
US20160375126A1 (en) 2007-02-02 2016-12-29 Baylor Research Institute Agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (dc-asgpr)
US20120282281A1 (en) 2007-02-02 2012-11-08 Baylor Research Insitute Agents that Engage Antigen-Presenting Cells Through Dendritic Cell Asialoglycoprotein Receptor (DC-ASGPR)
US8449888B2 (en) 2007-02-02 2013-05-28 Baylor Research Institute Vaccines based on targeting antigen to DCIR expressed on antigen-presenting cells
US7786267B2 (en) 2007-02-02 2010-08-31 Baylor Research Institute Multivariable antigens complexed with targeting humanized monoclonal antibody
US9453074B2 (en) 2007-02-02 2016-09-27 Baylor Research Institute Agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (DC-ASGPR)
US20080254044A1 (en) 2007-02-02 2008-10-16 Baylor Research Institute Multivariable Antigens Complexed with Targeting Humanized Monoclonal Antibody
US8236934B2 (en) 2007-02-02 2012-08-07 Baylore Research Institute Agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (DC-ASGPR)
WO2008097817A2 (en) 2007-02-02 2008-08-14 Baylor Research Institute Multivariable antigens complexed with targeting humanized monoclonal antibody
US20120237513A1 (en) 2007-02-02 2012-09-20 Baylor Research Institute Vaccines Based on Targeting Antigen to DCIR Expressed on Antigen-Presenting Cells
US8728481B2 (en) 2007-02-02 2014-05-20 Baylor Research Institute Agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (DC-ASGPR)
US9339556B2 (en) 2007-02-23 2016-05-17 Baylor Research Institute Activation of human antigen-presenting cells through dendritic cell lectin-like oxidized LDL receptor-1 (LOX-1)
US20080233140A1 (en) 2007-02-23 2008-09-25 Baylor Research Institute Therapeutic Applications of Activation of Human Antigen-Presenting Cells Through Dectin-1
US8481314B2 (en) 2007-02-23 2013-07-09 Baylor Research Institute Activation of human antigen-presenting cells through dendritic cell lectin-like oxidized LDL receptor-1 (LOX-1)
US20120039916A1 (en) 2010-08-13 2012-02-16 Baylor Research Institute Novel vaccine adjuvants based on targeting adjuvants to antibodies directly to antigen-presenting cells
US20120121592A1 (en) 2010-10-13 2012-05-17 Baylor Research Institute Targeting Antigens to Human Dendritic Cells Via DC-Asialoglycoprotein Receptor to Produce IL-10 Regulatory T-Cells
US20120231023A1 (en) 2011-03-08 2012-09-13 Baylor Research Institute Novel Vaccine Adjuvants Based on Targeting Adjuvants to Antibodies Directly to Antigen-Presenting Cells
US20120244155A1 (en) 2011-03-22 2012-09-27 Baylor Research Institute Dendritic Cells (DCs) Targeting for Tuberculosis (TB) Vaccine
US20120301465A1 (en) 2011-03-25 2012-11-29 Institut National De La Sante Et De La Recherche Medicale (Inserm) Compositions and methods to immunize against hepatitis c virus
US20120315269A1 (en) 2011-05-05 2012-12-13 Baylor Research Institute Immunoglobulin-like transcript (ilt) receptors as cd8 antagonists

Non-Patent Citations (129)

* Cited by examiner, † Cited by third party
Title
Accession No. NP_498023, Nov. 2008, pp. 1-2.
Ahmad-Nejad, et al., "Bacterial CpG-DNA and Lipopolysaccharides Activate Toll-Like Receptors at Distinct Cellular Compartments," European J. lmmunol., (2002), 32:1958-1968.
Arce, Ignacio et al., "The human C-type lectin CLECSF18 is a novel monocyte/macrophage endocytic receptor", European Journal of Immunology, 2004, vol. 34(1 ), pp. 210-220.
Asea, A., et al., "HSP70 Stimulates Cytokine Production Through a CD14-Dependant Pathway, Demonstrating its Dual Role as a Chaperone and Cytokine," Nat Med, (2000), 6:435-442.
Balazs, F., et al., "Blood Dendritic Cells Interact with Splenic Marginal Zone B Cells to Initiate T-lndependent Immune Responses," Immunity, (2002), 17:341-352.
Balch, et al., J. Biol Chem. 273(29): 18656-64, 1998.
Banchereau J., et al., "Immune and Clinical Responses in Patients with Metastatic Melanoma to CD34(+) Progenitor-Derived Dendritic Cell Vaccine," Cancer Res, (2001), 61(17):6451-8.
Banchereau, J., et al., "Dendritic Cells and the Control of Immunity," Nature, (1998), 392:245-252.
Banchereau, J., et al., "Dendritic Cells as Vectors for Thereapy," Cell, (2001), 106(3):271-4.
Banchereau, J., et al., "lmmunobiology of Dendritic Cells," Annu Rev lmmunol, (2000), 18:767-811.
Barak, Y. et al., "Matching Fusion Protein Systems for Affinity Analysis of Two Interacting Families of Proteins: the Cohesin-Dockerin Interaction", J. Mo. Recognit, 2005:18:491-501.
Barrios, et al., "Length of the Antibody Heavy Chain Complementarity Determining Region 3 as a Specificity—Determining Factor", J. Mol. Recognit. 2004: 17:332-338.
Bates, E. E., et al., "APCs Express DCIR, a Novel C-Type Lectin Surface Receptor Containing an lmmunoreceptor Tyrosine-Based Inhibitory Motif," J lmmunol, (1999), 163(4):1973-83.
Bausinger, H., et al., "Endotoxin-Free Heat-Shock Protein 70 Fails to Induce APC Activation," Eur J lmmunol (2002), 32:3708-3713.
Bayer, et al., Trends Biotechnol. 12: 379-386, 1994.
Bendsten, J.D., et al., "Improved Prediction of Signal Peptides: SignaiP 3.0." J Mol Bioi, (2004), 340(4):783-95.
Berard, F., et al., "Cross-Priming of Naive CDS T Cells Against Melanoma Antigens Using Dendritic Cells Loaded with Killed Allogeneic Melanoma Cells," J Exp Med, (Dec. 2000), 192(11):1535-44.
Bergtold, A., et al., "Cell Surface Recycling of Internalized Antigen Permits Dendritic Cell Priming of B Cells," Immunity (2005), 23:503-514.
Bernasconi, N. L., et al., "Maintenance of Serological Memory by Polyclonal Activation of Human Memory B Cells," Science (2002), 298:2199-2202.
Beutler, B., et al., "Genetic Analysis of Host Resistance: Toll-Like Receptor Signaling and Immunity at Large," Annu Rev lmmunol (2006), 24:353-389.
BIOLOGICAL ABSTRACTS, 1 November 2007, Philadelphia, PA, US; GRUENEBACH FRANK, ET AL: "hDectin-1 mediates cross-presentation of epitopes via the cytosolic pathway" XP002563205
Bonifaz, L.C., et al., "In Vivo Targeting of Antigens to Maturing Dendritic Cells via the DEC-205 Receptor Improves T Cell Caccination" J Exp Med, (Mar. 2004), 199(6):815-24.
Bonifaz, Laura, et al., "Efficient Targeting of Protein Antigen to the Dendritic Cell Receptor DEC-205 in the Steady 8 State Leads to Antigen Presentation on Major Histocompatibility Complex Class I Products and Peripheral CD8 T Cell D Tolerance," J. Exp. Med., Dec. 16, 2002, vol. 196, No. 12, pp. 1627-1638.
Brown, G. D., "Dectin-1: a signalling non-TLR pattern-recognition receptor," Nat Rev lmmunol (2006), 6:33-43.
Burgdorf et al., J Immunol. 176:6770-6, 2006.
Cambi, A., et al., "The C-type lectin DC-SIGN (CD209) is an antigenuptake receptor for Candida albicans on dendritic cells," Eur J lmmunol (2003), 33:532-538.
Carter, et al., "Preferential Induction of CD4+ T Cell Responses through In Vivo Targeting of Antigen to Dendritic Cell-Associated C-Type Lectin-1", J. of lmmunol.,vol. 177, No. 4, 2006, pp. 2276-2284.
Carvalho, A.L., et al., "Cellulosome Assembly Reveal by the Crystal Structure of the Cohesin-Dockerin Complex." Proc Natl Acad Sci USA, (2003), 100(24):13809-14.
Chu. et al., "CpG Oligodeoxynucleotides Down-Regulate Macrophage Class II MHC Antigen Processing", J lmmunol 1999:163:1188-1194.
Colonna, M., et al., "Molecular Characterization of Two Novel C-Type Lectin-Like Receptors, one of which is Selectively Expressed in Human Dendritic Cells," Eur J lmmunol (2000), 30:697-704.
Cooper, A.M., et al., "Mice Lacking Bioactive IL-12 Can Generate Protective, Antigen-Specific Cellular Responses to Mycobacterial Infection Only if the IL-12 p40 Subunit Is Present," J lmmunol (2002), 168:1322-1327.
Craig et al., 2006, J. Biotech. vol. 121: 165-173.
Craxton, A., et al., "Macrophage and Dendritic Cell-Dependent Regulation of Human B-Cell Proliferation Requires the TNF Family Ligand BAFF," Blood, (2003), 101:4464-4471.
Daly, et al., Anal Lett. 34:1799-1827, 2001.
Deineste, Y., G. et al., "Involvement of LOX-1 in Dendritic Cell-Mediated Antigen Cross-Presentation." Immunity (2002), 17(3):353-62.
Deyev, S.M. et al., "Design of multivalent complexes using the barnase-barstar module", Nature Biotechnology, vol. 21, No. 12, Dec. 2003, pp. 1486-1492.
Ding, et al., "A Scaffoldin of the Bacteriodes cellulosolvens Celluslosome That Contains 11 Type II Cohesins", J. Bacteriology 182(17):4915-4925, Sep. 2000.
Ding, et al., "The Bacterial Scaffoldin: Structure, Function and Potential Applications in the Nanosciences", Genetic Engineering, vol. 25:209-225, (2003).
D'Ostiani, C. F., et al., "Dendritic Cells Discriminate between Yeasts and Hyphae of the Fungus Candida albicans: Implications for Initiation ofT Helper Cell Immunity In Vitro and In Vivo," J Exp Med {2000), 191:1661-1673.
Dubois, B., et al., "Dendritic cells directly modulate B cell growth and differentiation," J Leukoc Bioi (1999), 66:224-230.
Engering, et al., Trends Immunol. 23(10): 480-5, 2002.
Extended European Search Report for Application No. 08714180.0, dated Sep. 29, 2011, 14 pages.
Extended European Search Report for Application No. 08728767.8, dated Aug. 24, 2010, 11 pages.
Extended European Search Report for Application No. 08799678.1, dated Feb. 5, 2010, 12 pages.
Fierobe, H-P et al., "Design and Production of Active Cellulosome Chimeras," J Bioi Chem (2001 ), 276:21257-21261.
Figdor, C. G., et al., "C-Type Lectin Receptors on Dendritic Cells and Langerhans Cells," Nat Rev lmmunol, (2002), 2{2): 77-84.
Flamar, A.L. et al., "Antibody and Antigen Complexes Based on Cohesin and Dockerin Interaction are Versatile Tools for Eliciting and Monitoring Specific Immune Responses", Clinical Immunology, US Lnkd-DOI:1 0.1016/J. D Clim.2009.03.019, vol. 131, Jan. 1, 2009, pp. S9.
Flomes, L.M. et al., "Identification of Lectin-Like Receptors Expressed by Antigen Presenting Cells and Neutrophils and their Mapping to a Novel Gene Complex," Immunogenetics, (2004), vol. 56, pp. 506-517.
Fradin, C., et al., "˜-1 ,2-Linked Oligomannosides from Candida albicans Bind to a 32-Kilodalton Macrophage Membrane Protein Homologous to the Mammalian Lectin Galectin-3," Infect lmmun (2000), 68:4391-4398.
Frankel, A. F., "Increased Sophistication of lmmunotoxins," Clin Can Res, {2002), 8:942-944.
Gantner, B. N., et al., "Collaborative Induction of Inflammatory Responses by Dectin-1 and Toll-like Receptor 2," J Exp Med (2003), 197:1107-1117.
Geijtenbeek, T. B., et al., "DC-SIGN-ICAM-2 Interaction Mediates Dendritic Cell Trafficking," Nat lmmunol {2000), 1:353-357.
Geijtenbeek, T. B., et al., "Identification of DC-SIGN, a Novel Dendritic Cell-Specific ICAM-3 Receptor that Supports Primary Immune Responses," Cell (2000), 100:575-585.
Geijtenbeek, T. B., et al., "Mycobacteria Target DC-SIGN to Suppress Dendritic Cell Function," J Exp Med (2003), 197:7-17.
Geijtenbeek, T.B., et al., "Self-and Nonself-Recognition by C-type Lectins on Dendritic Cells." Annu Rev lmmunol (2004), 22:33-54.
Gross, J. A., et al., "TACI and BCMA are Receptors for a TNF Homologue Implicated in B-cell Autoimmune Disease," Nature (2000), 404:995-999.
Grueneback, et al., "Human {h) Dectin-1 is a Member of C-Type-Lectin-Like Receptor Family that was Shown to be the Major Receptor for Fungal Beta-Giucans . . . " Abstract from 49th Annual Meeting of the American Society of Dematology, Dec. 2007, XP002563205, 2 pages.
Hayashida, K., et al., "Lectin-Like Oxidized LDL Receptor-1 (LOX-1) Supports Adhesion of Mononuclear Leukocytes and a Monocyte-Like Cell Line THP-1 Cells Under Static and Flow Conditions," FEBS Letters, (2002), 511:133-138.
Huang, et al., "Cloning and Characterization of a Novel ITIM Containing Lectin-like lmmunoreceptor LLIR and Its Two Transmembrane Region Deletion Variants", Biochem. Biophys. Res. Commun., 281(1):131-140, (2001).
International search Report and Written Opinion for PCT/US08/52865, dated Aug. 13, 2008, 8 pages.
International Search Report and Written Opinion for PCT/US2008/052714, dated Jul. 8, 2008, 8 pages.
International Search Report and Written Opinion for PCT/US2008/054785, dated Sep. 25, 2008, 12 pages.
International Search Report and Written Opinion for PCT/US2008/054792, dated Aug. 13, 2008, 10 pages.
International Search Report and Written Opinion for PCT/US2008/054798, dated Dec. 8, 2008, 13 pages.
International Search Report and Written Opinion for PCT/US2008/52850, dated Sep. 24, 2008, 9 pages.
Janeway, et al., Immunobiol. 3:3-3: 4, 1997.
Jeannin, P., et al., "Complexity and Complementarity of Outer Membrane Protein A Recognition by Cellular and Humoral Innate Immunity Receptors," Immunity (2005), 22:551-560.
Jego, G., et al., "Plasmacytoid Dendritic Cells Induce Plasma Cell Differentiation through Type I Interferon and lnterleukin 6," Immunity (2003), 19:225-234.
Jiang, W., et al., Nature, 375, pp. 151-155, May 11, 1995.
Jindou, S. et al., "Interaction between a Type-11 Dockerin Domain and a Type-11 Cohesin Domain from Clostridium thermocellum Cellulosome", Biosci. Biotechnol. Biochem. 68(4):924-926,(2004).
Kakutani, M., et al., "A platelet-endothelium interaction mediated by lectin-like oxidized low-density lipoprotein receptor-1," PNAS (2000), 97:360-364.
Karni et al., Journal of Immunology vol. 177: 4196-4202, 2006.
Karpol, A. et al., "Engineering a Reversible, High-Affinity System for Efficient Protein Purification Based on the Cohesin-Dockerin Interaction", J. Mol. Recognit. 2009, vol. 22:91-98.
Kikuchi, T., et al., "Dendritic Cells Genetically Modified to Express CD40 Ligand and Pulsed with Antigen can Initiate Antigen-Specific Humoral Immunity Independent of CD4+ T Cells," Nat Med, (2000), 6:1154-1159.
Klimka et al., "Human Anti-CD30 Recombinant Antibodies by Guided Phage Antibody Selection Using Cell Planning", Brit. J. Cancer, vol. 83, 252-260, 2000.
Kobrin, et al., "A V Region Mutation in a Phosphocholine-Bindng Monoclonal Antibody Results in Loss of Antigen Binding", J. Immunology, 146:2017-2020 (1991).
Latz, et al., "TLR9 Signals After Translocating from the ER to CpG DNA in the Lysosome", Nat. lmmunol., (2004), vol. 5:190-198.
Lee, S. J., et al., "Mannose Receptor-Mediated Regulation of Serum Glycoprotein Homeostasis," Science (2002), 295:1898-1901.
Li, et al., "Targeting Self and Foreign Antigens to Dendritic Cells via DC-ASGPR Generates IL-10-Producing Suppressive CD4+T Cells," J. Exp. Med., 209: 109-121, 2012.
Li, et al., Clin Exp Immunol. 162:251-61, 2010.
Luft, et al., International Immunology 14(4):367-380, 2002.
Ma, "Genome-Wide Analysis of Human Peripheral Leukocyte Gene Expression," PhD Thesis, p. 51 & 60, 2003.
MacLennan, et al., "Dendritic Cells, BAFF, and APRIL: Innate Players in Adaptive Antibody Responses," Immunity (2002), 17:235-238.
Maeda, N., et al., "The Cell Sulface Receptor DC-SIGN Discriminates between Mycobacterium Species through Selective Recognition of the Mannose Caps on Lipoarabinomannan," J Bioi Chern, (2003), 278:5513-5513.
Marks et al., "By-Passing Immunization: Human Antibodies from V-gene Libraries Displayed on Phage", J. Mol. Bioi., (1991), 222:581-597.
Mechaly, A. et al., "Cohesin-Dockerin Interaction in Cellulosome Assembly", J. Biological Chemistry, Mar. 30, 2001, vol. 276, No. 13, pp. 9883-9888.
Mehta et al., "Lectin-like, oxidized low-density lipoprotein receptor-1 (LOX-1): a critical player in the development of aterosclerosis and related disorders", Cardiovasc. Res., 69:36-45, 2006.
Mellman, I. et al., "Dendritic Cells: Specialized and Regulated Antigen Processing Machines," Cell, (2001), 106 (3):255-8.
Moore, P. A., et al., "BLyS: Member of the Tumor Necrosis Factor Family and B Lymphocyte Stimulator," Science (1999), 285:260-263.
Moriwaki, H., et al., "Expression of Lectin-Like Oxidized Low Density Lipoprotein Receptor-1 in Human and Murine Macrophages: Upregulated Expression by TN F-a," FEBS Letters, (1998), 440:29-32.
Neidhardt-Berard, E.M., et al., "Dendritic Cells Loaded with Killed Breast Cancer Cells Induce Differentiation of Tumor-Specific Cytotoxic T Lymphocytes" Breast Cancer Res, (2004), 6(4):R322-8.
Netea, M.G., et al., "CD40/CD40 Ligand Interactions in the Host Defense Against Disseminated Candida Albicans Infection: the Role of Macrophage-Derived Nitric Oxide," Eur J lmmunol, (2002), 32:1455-1463.
Office Communication in Israeli Patent Application No. 224539 dated Apr. 23, 2013.
Palucka, A.K., et al., "Human Dendritic Cell Subsets in NOD/SCID Mice Engrafted with CD34+ Hematopoieticprogenitors," Blood, (2003), 102(9):3302-1 0.
Proudfoot et al., "Receptor-Mediated Delivery of Antigens to Dendritic Cells: Anticancer Applications", Molec. Pharm., vol. 4, 2007, pp. 58-72.
Pyz, E., et al., "C-Type Lectin-Like Receptors on Myeloid Cells," Ann Med, (2006), 38:242-251.
Q1, H., et al., "Extrafollicular Activation of Lymph Node B Cells by Antigen-Bearing Dendritic Cells," Science (2006), 312:1672-1676.
Ramakrishna, et al., "Toll-like Receptor Activation Enhances Cell-Mediated Immunity Inducted by an Antibody Vaccine Targeting Human Dendritic Cells", Journal of Translational Medicine, vol. 5:5, pp. 1-14, Jan. 2007.
Ramakrishna, V., et al., "Mannose Receptor Targeting of Tumor Antigen PMEL 17 to Human Dendritic Cells Directs Antimelanoma T Cell Responses via Multiple HLA Molecules" J lmmunol, (2004), 172(5):2845-52.
Reddy, M.P., et al., "Elimination of FC Receptor-Dependent Effector Functions of a Modified lgG4 Monoclonal Antibody to Human CD4," J lmmunol ,(2000), 164(4):1925-33.
Riemer, et al., Mol Immunol. 42: 1121-4, 2005.
Rocca-Serra, et al., Nature. 304:353-355, 1983.
Rose, et al., J. Immunol. MRC1 mannose receptor, C type 1 [Homo sapiens], 1999, http://www.ncbi.nlm.nih.gov/gene/4360.
Rossi, E.A. et al., "Development of New Multivalent-bispecific Agents for Pretargeting Tumor Localization and Therapy", Clinical Cancer Research, vol. 9, Sep. 1, 2003, pp. 3886S-3896S.
Ruprecht, C. R., et al., "Toll-like Receptor Stimulation as a Third Signal Required for Activation of Human Naive B Cells," Eur J lmmunol, (2006), 36:810-816.
Saeland, S., "Langerin and the DC Asialoglycoprotein Receptor: Two Closely Related Endocytic Type-11 Lectins with Divergent Functions in Dendritic Cells," Journal of Investigative Dermatology, vol. 117, No. 4, Oct. 2001, p. 1003.
Sagoo, et al., "Regulatory T Cells and Therapeutic Cells," Curr. Opn. Organ Transplant, 13: 645-653, 2008.
Schaft, Neils, et al., "Dendritic Cell Vaccination and Other Strategies to Tip the Balance of the Immune System," Cancer lmmunol. lmmunother, (2008), 57:913-928.
Shortman, Ken, et al., "Mouse and Human Dendritic Cell Subtypes," Nature Reviews, Immunology, vol. 2, Mar. 2002, pp. 454-465.
Smith, Daniel C., et al., "Lack of Dendritic Cell Maturation by the Plant Toxin Ricin," European J. lmmunol., (2004), 34:2149-2157.
Spooner, RA, et al., "Expression of Immunoglobulin Heavy Chain-Ricin a Chain Fusions in Mammalian Cells," Mol. lmmunol., Feb. 1994, 31(2):117-125.
Stancovski, et al., PNAS. 88:8691-5, 1991.
Starovasnik, Melissa A., et al., "Structural Mimicry of a Native Protein by a Minimized Binding Domain," Proc. Nat. Acad. Sci. USA, Sep. 1997, vol. 94, pp. 10080-10085.
Steinman, "In vitro production of antigen-activated dendritic cell precursor for use as an immunogen in a vaccine; application in juvenile diabetes, multiple sclerosis, myasrhenia gravis and atopic dermatitis therapy", Vaccine, 12(5):478, 1994.
Steinman, Ralph M., et al., "Active Immunization Against Cancer with Dendritic Cells: The Near Future," Int. J. Cancer, {2001), 94, 459-473.
Tacken, Paul J., et al., "Effective Induction of Naive and Recall T-Cell Responses by Targeting Antigen to Human Dendritic Cells via a Humanized Anti-DC-SIGN Antibody," Blood, May 5, 2005, 106:1278-1285.
Tailleux, L., et al., "DC-SIGN Is the Major Mycobacterium tuberculosis Receptor on Human Dendritic Cells," J Exp Med (2003), 197:121-127.
Tan, et al., "Mannose Receptor-Mediated Uptake of Antigens Strongly Enhances HLA Class 11-Restricted Antigen Presentation by Cultured Dendritic Cells," Eur. J. lmmunol., 1997, 27:2426-2435.
Trombetia, E. Sergio, et al., "Cell Biology of Antigen Processing in Vitro and In Vivo," Annu. Rev. lmmunol., 2005, 23:975-1028.
Trumpfheller, Christine, et al., "Intensified and Protective CD4+ T Cell Immunity in Mice with Anti-Dendritic Cell HIV Gag Fusion Antibody Vaccine," Mar. 20, 2006, vol. 203, No. 3, pp. 607-617.
Valladeau, J., et al., "Immature Human Dendritic Cells Express Asialoglycoprotein Receptor lsoforms for Efficient Receptor-Mediated Endocytosis," J lmmunol (2001), 167:5767-5774.
Wang, Y., et al., "CD40 Is a Cellular Receptor Mediating Mycobacterial Heat Shock Protein 70 Stimulation of CC-Chemokines," Immunity (2001), 15:971-983.
Wiley, K.N., et al., "The In-Vitro Inhibition of Rat Alloantigen Presentation by lmmunotoxins—IMplications for Allografting," Clin. Exp. lmmunol., (1989), 76:132-137.
Wykes, M. N., et al., "Dendritic cell-8-cell interaction: dendritic cells provide B cells with CD40-independent proliferation signals and CD40-dependent survival signals," Immunology (2000), 100:1-3.
Wykes, M. N., et al., "Dendritic cells and follicular dendritic cells express a novel ligand for CD38 which influences their maturation and antibody responses," Immunology (2004), 113:318-327.
Xie et al., "Generation of Anti-LOX-1 Cytotoxic T Lymphocytes by AAV Manipulation of Dendritic Cells towards Preventing Atheosclerosis", Mol. Ther., 11(Suppl. 1):S362-S363, 2005.
Zhu, Zhenping, et al., "Inhibition of Tumor Growlh and Metastasis by Targeting Rumor-Associated Angiogenesis with Antagonists to the Receptors of Vascular Endothelial Growlh Factor," Investigational New Drugs, 1999, 17:195-212.
Zhu, Zhenping, et al., "Inhibition of Tumor Growth and Metastasis by Targeting Rumor-Associated Angiogenesis with Antagonists to the Receptors of Vascular Endothelial Growth Factor," Investigational New Drugs, 1999, 17:195-212.
Zymosan, Dectin-1 Review, "A Major Receptor in Antifungal Immunity" 2008.

Also Published As

Publication number Publication date
EP2115129A4 (en) 2011-11-02
TWI422594B (en) 2014-01-11
CA2715045C (en) 2020-08-04
US9453074B2 (en) 2016-09-27
IL200093A0 (en) 2010-04-15
US20080206262A1 (en) 2008-08-28
TW200846370A (en) 2008-12-01
WO2008097870A3 (en) 2010-01-07
JP2010518024A (en) 2010-05-27
US20190231865A1 (en) 2019-08-01
WO2008097870A2 (en) 2008-08-14
NZ593639A (en) 2012-08-31
EP2115129B1 (en) 2013-11-13
NZ578756A (en) 2012-02-24
US20140227268A1 (en) 2014-08-14
AU2008214036A1 (en) 2008-08-14
US8728481B2 (en) 2014-05-20
EP2115129A2 (en) 2009-11-11
KR20090114432A (en) 2009-11-03
US20160375126A1 (en) 2016-12-29
CA2715045A1 (en) 2008-08-14
IL218085A0 (en) 2012-03-29
US10279030B2 (en) 2019-05-07
US20120282281A1 (en) 2012-11-08
CN101952414A (en) 2011-01-19
ZA200905274B (en) 2010-04-28
US20220152192A1 (en) 2022-05-19
AU2008214036B2 (en) 2012-11-01
MX2009008211A (en) 2009-08-12
US8236934B2 (en) 2012-08-07
BRPI0807152A2 (en) 2014-04-22

Similar Documents

Publication Publication Date Title
US11173202B2 (en) Agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (DC-ASGPR)
EP2522718B1 (en) Therapeutic applications of activation of human antigen-presenting cells through dectin-1
US9234040B2 (en) Vaccines based on targeting antigen to DCIR expressed on antigen-presenting cells
US9339556B2 (en) Activation of human antigen-presenting cells through dendritic cell lectin-like oxidized LDL receptor-1 (LOX-1)
US20080254047A1 (en) Activation of Human Antigen-Presenting Cells Through CLEC-6
AU2012245170B2 (en) Agents that engage antigen - presenting cells through dendritic cell asialoglycoprotein receptor (dc-asgpr)
AU2014240262B2 (en) Agents that engage antigen - presenting cells through dendritic cell asialoglycoprotein receptor (dc-asgpr)
BANCHEREAU et al. Sommaire du brevet 2715045
BANCHEREAU et al. Patent 2715045 Summary

Legal Events

Date Code Title Description
AS Assignment

Owner name: BAYLOR RESEARCH INSTITUTE, TEXAS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:OH, SANGKON;LI, DAPENG;REEL/FRAME:048767/0316

Effective date: 20070306

FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO SMALL (ORIGINAL EVENT CODE: SMAL); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE AFTER FINAL ACTION FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STPP Information on status: patent application and granting procedure in general

Free format text: PUBLICATIONS -- ISSUE FEE PAYMENT VERIFIED

STCF Information on status: patent grant

Free format text: PATENTED CASE

CC Certificate of correction